IL297073B2 - Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens - Google Patents

Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens

Info

Publication number
IL297073B2
IL297073B2 IL297073A IL29707322A IL297073B2 IL 297073 B2 IL297073 B2 IL 297073B2 IL 297073 A IL297073 A IL 297073A IL 29707322 A IL29707322 A IL 29707322A IL 297073 B2 IL297073 B2 IL 297073B2
Authority
IL
Israel
Prior art keywords
seq
uniprotkb
lamp
cell
cells
Prior art date
Application number
IL297073A
Other languages
Hebrew (he)
Other versions
IL297073A (en
IL297073B1 (en
Original Assignee
Immunomic Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunomic Therapeutics Inc filed Critical Immunomic Therapeutics Inc
Publication of IL297073A publication Critical patent/IL297073A/en
Publication of IL297073B1 publication Critical patent/IL297073B1/en
Publication of IL297073B2 publication Critical patent/IL297073B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/64Medicinal preparations containing antigens or antibodies characterised by the architecture of the carrier-antigen complex, e.g. repetition of carrier-antigen units
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Description

WO 2018/204534 PCT/US2018/030725 LAMP (LYSOSOMAL ASSOCIATED MEMBRANE PROTEIN) CONSTRUCTS COMPRISING CANCER ANTIGENS Field of the Invention [0001] The invention relates to improved LAMP Constructs comprising cancer antigens and their use in treating subjects suffering from hyperproliferative disorders and/or cancer. Prime boost protocols utilizing the improved LAMP Constructs described herein are also described.
Discussion of the Related Art [0002] In the following discussion, certain articles and methods will be described for background and introductory purposes. Nothing contained herein is to be construed as an "admission" of prior art. Applicant expressly reserves the right to demonstrate, where appropriate, that the articles and methods referenced herein do not constitute prior art under the applicable statutory provisions.[0003] DNA vaccines are new and promising candidates for the development of both prophylactic and therapeutic vaccines. They are proven to be safe and the lack of immune responses to a vector backbone may be a definitive advantage if repetitive cycles of vaccination are required to achieve clinical benefits. However, one perceived disadvantage of conventional DNA vaccines is their low immunogenicity in humans. A key limiting step in the immunogenicity of epitope-based DNA vaccines may be the access of epitopes to the MHCII presentation pathway to T cells, which is likely a stochastic process in the case of a vaccine without targeting technology.[0004] U.S. Pat. No. 5,633,234 describes chimeric proteins comprising an antigenic domain of modified influenza hemagglutinin (HA) and a cytoplasmic endosomal/lysosomal targeting signal which effectively target antigens to that compartment. The antigenic domain was processed and peptides from it presented on the cell surface in association with major histocompatibility (MHC) class II molecules. The cytoplasmic tail of LAMP-1 was used to form the endosomal/lysosomal targeting domain of the chimeric protein.[0005] U.S. Pat. No. 8,318,173 extended these initial observations to describe chimeric proteins (and the corresponding DNAs that encode these proteins) comprising the HIV-1 Gag protein inserted between the full lumenal domain and a transmembrane domain of LAMP-1. This construct was introduced into dendritic cells which were then reported to target the MHC II pathway.[0006] This approach has proved useful in increasing cellular and humoral responses to several virus antigens, human papillomavirus E7, dengue virus membrane protein, HIV-1 gpl60 membrane protein, HIV-p55 Gag, West Nile membrane protein, hepatitis C virus NS3 protein and cytomegalovirus pp65 (see, e.g., Bonini, et al., J. Immunol. 166: 5250-5257, 2001). The enhanced immune response can be attributed to co- localization of LAMP with MHC II and the more efficient processing and delivery of antigenic peptides. In addition, LAMP-targeting is reported to result in the presentation of an increased number of immunogenic WO 2018/204534 PCT/US2018/030725 epitopes, thus inducing a qualitatively broadened immune response compared to untargeted antigen. For example, Fernandes et al., 2000, Fur. J. Immunol. 30(8): 2333-43, demonstrated an increase in the number of presented peptides of a LAMP-trafficked OVA antigen encoded in a vaccinia vector. Of 12 peptides generated from exogenously supplied OVA, 9 were presented by an OVA/LAMP chimera, as compared to only 2 by the construct without LAMP.[0007] While it has been determined that the cytoplasmic domain of LAMP is necessary (in conjunctionwith a signal sequence and transmembrane domain), it is not always sufficient for endosomal/lysosomal trafficking of all antigens. Instead, the full lumenal domain of LAMP has been shown to be also required for the trafficking of proteins to the lysosomal vesicular pathway.[0008] However, even with the presence of the complete lumenal domain and the complete transmembrane/cytoplasmic tail of LAMP ("complete LAMP Constructs"), it has increasingly been found that the efficacy of a particular antigen to raise an immune response is highly dependent on the particular sequence used in these constructs. In fact, different antigenic fragments of the same protein when inserted into the complete LAMP constructs have been found to not elicit the same immune response. Sometimes the antigen fragment generates an immune response and other times it does not. These observations make the ability to predict ahead of time which particular antigenic sequence from a protein of interest will raise an immune response difficult with the complete LAMP Constructs.[0009] Moreover, in generating the complete LAMP Constructs, it has been repeatedly observed that the full lumenal domain is required to properly express and process an antigen. For example, in Godinho et al., PLoS ONE 9(6): 9(6): 699887. doi:10.1371/journal.pone.0099887, the authors reported that the complete and intact lumenal domain was the necessary minimal region needed to target an antigen to the lysosomes and that fragments of the lumenal domain did not work. See, id. at page 6.[0010] Specifically, the Godinho authors showed that by completely removing the first luminal domain and some of the second luminal domain (i.e., Tl-Lum/gag construct), both protein expression and antibody response is decreased. Similarly, removing 25% of first luminal domain but having an intact second luminal domain (i.e., T2-lum/gag), both protein expression and antibody response comparatively increased but still less than the results obtained with the complete LAMP construct.[0011] Moreover, the authors acknowledged that the ability to raise an immune response is dependent upon the particular antigen and the epitopes used in these complete LAMP Constructs. For example, on page 9, column 2, the authors state "accordingly, previous studies demonstrated that DNA vaccines that generate Gag secreted as VLP, or in a soluble form, induce different levels of T and B cell activation, which were also different from the response induced by cytoplasmic Gag." Moreover, insertion of an antigenic sequence between the full lumenal domain of LAMP and the full transmembrane/cytoplasmic domain of LAMP as has been described in the literature can result in such large polynucleotide sequences that it can become either too costly to produce at commercial levels or impractical from a scientific perspective.
WO 2018/204534 PCT/US2018/030725 id="p-12" id="p-12" id="p-12"
[0012] Thus, there is a need to design new and improved LAMP Constructs that can be used as vaccines to effectively treat, for example, hyperproliferative disorders and/or cancer. Moreover, once improved, these new LAMP Constructs can be used to generate antibodies.
SUMMARY OF THE INVENTION [0013] This Summary is provided to introduce a selection of concepts in a simplified form that are further described below in the Detailed Description. This Summary is not intended to identify key or essential features of the claimed subject matter, nor is it intended to be used to limit the scope of the claimed subject matter. Other features, details, utilities, and advantages of the claimed subject matter will be apparent from the following written Detailed Description including those aspects illustrated in the accompanying drawings and defined in the appended claims.[0014] It is an object of this invention to provide novel constructs ("improved LAMP Constructs") comprising specific fragments and/or variants of LAMP domains that effectively present the cancer antigens specified herein to the immune system to generate an enhanced immune response. These improved LAMP Constructs effectively direct the antigens to the lysosomal/endosomal compartment where they are processed and presented to major histocompatibility complex (MHC) class II molecules so that helper T cells are preferentially stimulated and/or antibodies are generated.[0015] The improved LAMP Constructs and methods described herein may elicit an immune response in a subject. The immune response may be an immune response to the epitopes of the antigens in the improved LAMP Construct (e.g., vaccine). Vaccines arm the immune system of the subject such that the immune system may detect and destroy that which contains the antigens of the vaccines in the subject. The improved LAMP Constructs and methods described herein may elicit a Thl immune response in the subject. Thl immune responses may include secretion of inflammatory cytokines (e.g., IFNy, TNFa) by a subset of immune cells (e.g., antigen specific T-cells). In some cases, the inflammatory cytokines activate another subtype of immune cells (e.g., cytotoxic T-cells) which may destroy that which contains the antigen in the subject.[0016] In some cases, the epitopes and/or antigens used in the improved LAMP Constructs and methods described herein may be recognized by the immune system of a subject to elicit a Thl immune response and release Type I cytokines. The Thl response may be initiated by the interaction between the epitope and the T- cell, more specifically, the major histocompatibility complex (MHC) expressed by the T-cell. For example, high affinity binding of an epitope to an MHC receptor may stimulate a Thl response. MHC receptors may be at least one of a plurality of types of MHC receptors. The MHC receptors engaged on a T-cell may vary across individuals in a population.[0017] In some cases, the immune response is a Type 1 immune response. In some cases, the immune response is characterized by a ratio of Type I cytokine production to Type II cytokine production that is greater than 1. In some cases, the immune response is characterized by a ratio of Type I cytokine production to Type II WO 2018/204534 PCT/US2018/030725 cytokine production that is less than 1. In some cases, the immune response is characterized by a ratio of IFNy production to IL-10 production that is greater than 1. In some cases, the immune response is characterized by a ratio of IFNy production to IL-10 production that is less than 1.[0018] Prime boost protocols are also contemplated. For example, the invention further provides a method for generating an immune response in a subject to a cancer antigen, comprising priming the subject with an improved LAMP Construct comprising a cancer antigen as described herein followed by at least one boosting of the subject with the antigen or a related antigen (e.g., a second antigen derived from the same or highly similar protein sequence). Mixtures of antigens can be used in either or both the priming and the boosting step. Use of an improved LAMP Construct for the prime step followed by an antigen boost step has been shown to significantly produce higher titers, indicating the power of LAMP in enhancing antibody response.[0019] It is yet another object of this invention to provide improved methods of treatment for cancer and/or hyperproliferative disorders by eliciting an anti-tumor immune response through stimulation of helper T cells.[0020] The invention further provides a nucleic acid molecule encoding any of the improved LAMP Constructs comprising the cancer antigens described herein. The invention also provides an improved LAMP Construct comprising an antigen to generate antibodies. The improved LAMP Construct can comprise a nucleic acid wherein the nucleic acid molecule is operably linked to an expression control sequence. In one preferred aspect, the improved LAMP Construct is a vaccine vector, suitable for vaccinating a patient. In another aspect, the invention provides a delivery vehicle comprising the improved LAMP Construct for facilitating the introduction of the nucleic acid molecule encoding the antigen into a cell. The delivery vehicle may be lipid- based (e.g., a liposome formulation), viral-based (e.g., comprising viral proteins encapsulating the nucleic acid molecule), or cell-based.[0021] In preferred embodiments, the invention provides an injectable composition comprising an improved LAMP Construct comprising a cancer antigen of interest for eliciting an immune response (e.g., generation of antibodies) in a mammal to the cancer antigen. In preferred embodiments, this vaccine generates a preferential Thl response to a Th2 response. The improved LAMP Constructs comprise at least one epitope of a cancer antigen as described herein.[0022] The invention also provides a cell comprising any of the improved LAMP Constructs described herein. In one aspect, the cell is an antigen presenting cell. The antigen presenting cell may be a professional antigen presenting cell (e.g., a dendritic cell, macrophage, B cell, and the like) or an engineered antigen presenting cell (e.g., a non-professional antigen presenting cell engineered to express molecules required for antigen presentation, such as MHC class II molecules). The molecules required for antigen presentation may be derived from other cells, e.g., naturally occurring, or may themselves be engineered (e.g. mutated or modified to express desired properties, such as higher or lower affinity for an antigenic epitope). In one aspect, the antigen presenting cell does not express any co-stimulatory signals and the antigen is an auto-antigen.
WO 2018/204534 PCT/US2018/030725 id="p-23" id="p-23" id="p-23"
[0023] The invention additionally provides a kit comprising a plurality of cells comprising any of the improved LAMP Constructs described herein. At least two of the cells express different MHC class II molecules, and each cell comprises the same LAMP Construct. In one aspect, a kit is provided comprising an improved LAMP Construct and a cell for receiving the vector.[0024] The invention also provides a transgenic animal comprising at least one of the cells and/or at least one of the improved LAMP Constructs described herein. The invention also provides a transgenic animal comprising at least one of the cells described herein.[0025] The invention further provides a method for generating an immune response in an animal (e.g., a human or a non-human vertebrate) to an antigen, comprising: administering to the animal a cell as described above, wherein the cell expresses, or can be induced to express, the improved LAMP Construct in the animal. In one aspect, the cell comprises an MHC class II molecule compatible with MHC proteins of the animal, such that the animal does not generate an immune response against the MHC class II molecule. In one preferred aspect, the animal is a human.[0026] In one further aspect, the invention provides a method for eliciting an immune response to a cancer antigen, comprising administering to an animal, such as a human or a non-human vertebrate, any of the improved LAMP Constructs described herein. Preferably, the improved LAMP Construct is infectious for a cell of the animal. For example, the improved LAMP Construct may be a viral vector, such as a vaccinia improved LAMP Construct.[0027] For example, the invention further provides a method for generating an immune response in an animal to a cancer antigen, comprising priming the animal with an improved LAMP Construct comprising an antigen as described herein followed by at least one boosting of the animal. Use of an improved LAMP Construct for the prime step followed by an antigen boost step has been shown to significantly produce higher titers, indicating the power of LAMP in enhancing antibody response.[0028] In a further aspect, a cell is obtained from a patient, the improved LAMP Construct described herein is introduced into the cell and the cell or progeny of the cell is reintroduced into the patient. In one aspect, the cell is a stem cell-capable of differentiating into an antigen presenting cell. Treatments of human patients as well as veterinary use are specifically contemplated.[0029] The present invention also comprises methods of generating antibodies in a non-human vertebrate wherein the non-human vertebrate is injected with an improved LAMP Construct comprising an antigen of interest as described herein. The cancer antigen of interest is then efficiently presented to the immune system with the help of LAMP in the non-human vertebrate to raise antibodies against the antigen.[0030] Specifically, by combining presentation of the antigen of interest with LAMP, the antigen is then effectively transported to the cytoplasmic endosomal/lysosomal compartments, where the antigen can be processed and peptides from it presented on the cell surface in association with major histocompatibility (MHC) class II molecules.
WO 2018/204534 PCT/US2018/030725 id="p-31" id="p-31" id="p-31"
[0031] These generated antibodies can be isolated from the blood of the vertebrate (as polyclonals) andthen further isolated to generate monoclonal antibodies using standard techniques.[0032] In preferred embodiments, the genome of the non-human vertebrate comprises an introduced partially human immunoglobulin region, said introduced region comprising human immunoglobulin variable region locus coding sequences and non-coding sequences based on the endogenous immunoglobulin variable region locus of the non-human vertebrate. Preferably, non-human vertebrate’s genome has at least part or all of the endogenous immunoglobulin region removed.[0033] In further preferred embodiments, the production of human monoclonal antibodies in the non- human vertebrate requires that the host have at least one locus that will express human heavy chain immunoglobulin proteins and one locus that will express human light chain immunoglobulin proteins.[0034] In some aspects, the partially human immunoglobulin variable region locus comprises human Vh coding sequences and non-coding Vh sequences based on the endogenous Vh region of the non-human vertebrate. In these aspects, the partially human immunoglobulin variable region locus further comprises human D and J gene coding sequences and non-coding D and J gene sequences based on the endogenous genome of the non-human vertebrate host.[0035] In other aspects, the immunoglobulin region comprises an introduced region comprising human Vl coding sequences and non-coding Vl sequences based on the endogenous Vl region of the non-human vertebrate. More preferably, the introduced partially human immunoglobulin region comprising human Vl coding sequences further comprises human J gene coding sequences and non-coding J gene sequences based on the endogenous genome of the non-human vertebrate host.[0036] In certain aspects, the vertebrate is a mammal, and preferably the mammal is a rodent, e.g., a mouse or rat. In other aspects, the vertebrate is avian, e.g., a chicken. Other non-human vertebrates include rabbits, llamas, camels, a cow, a guinea pig, a hamster, a dog, a cat, a horse, a non-human primate, a simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon, rhesus macaque), or an ape (e.g. gorilla, chimpanzee, orangutan, gibbon).[0037] In further embodiments, the partially human immunoglobulin region comprises human Vh gene coding regions, and further comprises i) human D and J gene coding sequences and ii) non-coding D and J gene and pre-DJ sequences based on the endogenous genome of the non-human vertebrate host. In other aspects, the Vh gene coding regions derive (at least partially) from other sources—e.g., they could be rationally or otherwise designed sequences, sequences that are a combination of human and other designed sequences, or sequences from other species, such as nonhuman primates.[0038] In yet another specific aspect, the partially human immunoglobulin region comprises human Vl gene coding regions, and further comprises i) human J gene coding sequences and ii) non-coding J gene sequences based on the endogenous genome of the non-human vertebrate host. In a specific aspect, the partially human immunoglobulin region comprises human Vh coding regions, human D and J gene coding sequences, WO 2018/204534 PCT/US2018/030725 and non-coding D and J gene and pre-DJ sequences based on the endogenous genome of the non-human vertebrate host.[0039] The methods described herein can be used in the production and/or optimization of antibodies, including fully human antibodies, humanized antibodies, chimeric antibodies, for diagnostic and therapeutic uses. Hybridomas producing such antibodies are also a further object of the invention.[0040] These and other aspects, objects and features are described in more detail below.
BRIEF DESCRIPTION OF THE FIGURES [0041] The objects and features of the invention can be better understood with reference to the following detailed description and accompanying drawings.[0042] Figure 1illustrates the general scheme of different types of improved LAMP Constructs (identified as ILC-1, ILC-2, ILC-3, ILC-4, ILC-5 and ILC-6) that can be used as described herein.[0043] Figure 2Billustrates the domains of the LAMP proteins defined herein while Figure 2Adefines the specific amino acid boundaries of these domains for human LAMP-1 (SEQ ID NO: 1), human LAMP-2 (SEQ ID NO:2), human LAMP-3 (SEQ ID NO:3), human LIMP-2 (SEQ ID NO:4), human Endolyn (SEQ ID NO:5), human Macrosailin (SEQ ID NO:80), human LAMP-5 (SEQ ID NO:93) and human LIMBIC (SEQ ID NO:67). As described herein the LAMP lumenal domains, homology domains, transmembrane domains, the cytoplasmic tail and the signal sequences can be used to generate the improved LAMP Constructs ILC-1, ILC-2, ILC-3, ILC- 4, ILC-5 and ILC-6 as described herein.[0044] Figure 3provides alignment of LAMP-1 proteins found in other species as compared to human LAMP-1 (SEQ ID NO:1). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human LAMP-1 in Figures 2 and Figure 3 to the alignments shown in Figure 3.[0045] Figure 4provides alignment of LAMP-2 proteins found in other species as compared to human LAMP-2 (SEQ ID NO:2). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human LAMP-2 in Figures 2 and Figure 4 to the alignments shown in Figure 4.[0046] Figure 5provides alignment of LAMP-3 proteins found in other species as compared to human LAMP-3 (SEQ ID NO:3). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human LAMP-3 in Figures 2 and Figure 5 to the alignments shown in Figure 5.[0047] Figure 6provides alignment of LIMP-2 proteins found in other species as compared to human LIMP-2 (SEQ ID NO:4). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human LIMP-2 in Figures 2 and Figure 6 to the alignments shown in Figure 6. ר WO 2018/204534 PCT/US2018/030725 id="p-48" id="p-48" id="p-48"
[0048] Figure 7provides alignment of LIMBIC proteins found in other species as compared to human LIMBIC (SEQ ID NO:67). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human LIMBIC in Figures 2 and Figure 7 to the alignments shown in Figure 7.[0049] Figure 8provides alignment of Endolyn proteins found in other species as compared to human Endolyn (SEQ ID NO:5). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human Endolyn in Figures 2 and Figure 8 to the alignments shown in Figure 8.[0050] Figure 9provides alignment of Macrosailin proteins found in other species as compared to human Macrosailin (SEQ ID NO:80). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human Macrosailin in Figures 2 and Figure 9 to the alignments shown in Figure 9.[0051 ] Figure 10provides alignment of LAMP-5 proteins found in other species as compared to humanLAMP-5 (SEQ ID NO:93). The equivalent domains of these other species can be used to generate the improved LAMP Constructs described herein and are readily identifiable by comparing the domains identified for human LAMP-5 in Figures 2 and Figure 10 to the alignments shown in Figure 10.[0052] Figure 11shows results obtained when mice were immunized with HVEM-LAMP, HVEM, or LAMP on day 0, 7, and 14. On day 28, mice were bled and serum samples were isolated. HVEM specific IgG was examined by ELISA. Data represent geometric mean of antibody titers ± geometric SD, n=6. ** p value <0.01[0053] Figure 12shows results obtained when mice were immunized with HVEM-LAMP, HVEM, or LAMP on day 0, 7, and 14. On day 35, mice were boosted with 5pg HVEM protein in the presence of alum adjuvant. Mice were bled on day 49 and serum samples were isolated. HVEM specific IgG was examined by ELISA. Data represent geometric mean of antibody titers ± geometric SD, n=6. *** p value <0.001; **** p value <0.0001.[0054] Figure 13shows that LAMP alters the binding affinity of epitopes in CRD3/4 of HVEM.[0055] Figure 14confirms protein expression of tested improved LAMP Constructs. In each of Figures14-28, the labels "complete LAMP Construct", ILC-1, ILC-2, ILC-3 and ILC-4 correspond to the constructs as depicted in Figure 1.[0056] Figure 15shows that the improved LAMP Constructs induce Thl effector T cells producing INFy.[0057] Figure 16shows a particular improved LAMP construct (e.g., ILC-4 as shown in Figure 1) elicited a significantly higher T cell response against all Survivin peptide pools.
WO 2018/204534 PCT/US2018/030725 id="p-58" id="p-58" id="p-58"
[0058] Figure 17shows that CD4 T cells are the major source of IFNy producing cells and that the improved LAMP Constructs demonstrate an increase in the CD4 effector memory cell population over the Complete LAMP construct.[0059] Figure 18shows that the improved LAMP Constructs produced stronger Survivin-specific total IgG response in BALB/c mice.[0060] Figures 19 and 20provides the amino acid sequence of the cancer antigens that can be cloned, either singularly or in combination as described in Table 1 and paragraph [150], into the LAMP Constructs described herein and in Figures 1-10. Furthermore, Figure 19 provides the amino acid sequence of Survivin- LAMP construct tested in the Examples. The signal sequence of each construct is depicted as lower case and underlined letters; the Survivin sequence is depicted in capitalized, white letters, shaded in black; the luminal domain is depicted in italics and capitalized letters and the transmembrane/cytoplasmic domain is depicted in capitalized letter and shaded in grey, and in ILC-4, the second homology domain is bolded. Additional amino acids (LE and EE) may be included as part of the cloning linkers.[0061] Figure 21shows that IGFBP2 (39-328)-ILC-l Construct induced significantly higher IFNy producing effector T cells at the dose of lOug IM/EP immunization.[0062] Figure 22shows that IGFBP2 (39-328)-ILC-l Construct induced IFNy and/or TNFa producing CD4+ and CD8+ effector memory T cells at the dose of lOug IM/EP immunization.[0063] Figure 23shows that IGFBP-2-specific IgG production in C57BL/6 albino mice immunized with IGFBP2 (39-328)-ILC-l Construct by IM/EP immunization.[0064] Figure 24shows that IFNy induction with both pp65 and gB peptides.[0065] Figure 25shows that total IgG (left) and IgG2a (right) antibodies titers in serum of immunizedmice against CMV-transfected cell lysate.[0066] Figure 26shows PSMA-ILC-1 LAMP Construct induced significantly higher IFNy producing effector T cells at the dose of 20ug ID/EP immunization.[0067] Figure 27shows PSMA-ILC-1 LAMP Construct induced IFNy producing CD4+ and CD8+ effector memory T cells at the dose of 20ug ID/EP immunization.[0068] Figure 28shows total IgG and IgG2A production in C57BL/6 mice immunized with PSMA- ILC-1 LAMP Construct by immunization via ID/EP.
DETAILED DESCRIPTION [0069] The invention provides improved LAMP Constructs which can be used to generate vaccines and/or used to raise antibodies. The improved LAMP Constructs can be used to modulate or enhance an immune response. In one preferred aspect, the invention provides a method for treating a patient with cancer or a hyperproliferative disorder by providing an improved LAMP Construct comprising one or more of the cancer WO 2018/204534 PCT/US2018/030725 antigens as described herein. The improved LAMP Constructs can also be used to raise antibodies in non- human vertebrates, and in preferably, non-human mammals.
DEFINITIONS [0070] The following definitions are provided for specific terms which are used in the following written description.[0071] As used in the specification and claims, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a cell" includes a plurality of cells, including mixtures thereof. The term "a nucleic acid molecule" includes a plurality of nucleic acid molecules.[0072] As used herein, the term "comprising" is intended to mean that the improved LAMP Constructs and methods include the recited elements, but do not exclude other elements. "Consisting essentially of", when used to define improved LAMP Constructs and methods, shall mean excluding other elements of any essential significance to the combination. Thus, an improved LAMP Construct consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. "Consisting of" shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the improved LAMP Constructs of this invention. Embodiments defined by each of these transition terms are within the scope of this invention.[0073] The term "about" or "approximately" means within an acceptable range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, e.g., the limitations of the measurement system. For example, "about" can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5 fold, and more preferably within 2 fold, of a value. Unless otherwise stated, the term 'about' means within an acceptable error range for the particular value, such as ± 1-20%, preferably ± 1- 10% and more preferably ±1-5%.[0074] Where a range of values is provided, it is understood that each intervening value, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention.[0075] As used herein, "the lysosomal/endosomal compartment" refers to membrane-bound acidic vacuoles containing LAMP molecules in the membrane, hydrolytic enzymes that function in antigen processing, WO 2018/204534 PCT/US2018/030725 and MHC class II molecules for antigen recognition and presentation. This compartment functions as a site for degradation of foreign materials internalized from the cell surface by any of a variety of mechanisms including endocytosis, phagocytosis and pinocytosis, and of intracellular material delivered to this compartment by specialized autolytic phenomena (de Duve, Eur. J. Biochem. 137: 391, 1983). The term "endosome" as used herein and in the claims encompasses a lysosome.[0076] As used herein, a "lysosome-related organelle" refers to any organelle which comprises lysosymes and includes, but is not limited to, MIIC, CIIV, melanosomes, secretory granules, lytic granules, platelet-dense granules, basophil granules, Birbeck granules, phagolysosomes, secretory lysosomes, and the like. Preferably, such an organelle lacks mannose 6-phosphate receptors and comprises LAMP, but may or may not comprise an MHC class II molecule. For reviews, see, e.g., Blott and Griffiths, Nature Reviews, Molecular Cell Biology, 2002; Dell'Angelica, et al., The FASEB Journal 14: 1265-1278, 2000.[0077] As used herein, the terms "polynucleotide" and "nucleic acid molecule" are used interchangeably to refer to polymeric forms of nucleotides of any length. The polynucleotides may contain deoxyribonucleotides, ribonucleotides, and/or their analogs. Nucleotides may have any three-dimensional structure, and may perform any function, known or unknown. The term "polynucleotide" includes, for example, single-, double-stranded and triple helical molecules, a gene or gene fragment, exons, introns, mRNA, tRNA, rRNA, ribozymes, antisense molecules, cDNA, recombinant polynucleotides, branched polynucleotides, aptamers, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A nucleic acid molecule may also comprise modified nucleic acid molecules (e.g., comprising modified bases, sugars, and/or internucleotide linkers).[0078] As used herein, the term "peptide" refers to a compound of two or more subunit amino acids, amino acid analogs, or peptidomimetics. The subunits may be linked by peptide bonds or by other bonds (e.g., as esters, ethers, and the like).[0079] As used herein, the term "amino acid" refers to either natural and/or unnatural or synthetic amino acids, including glycine and both D or L optical isomers, and amino acid analogs and peptidomimetics. A peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long (e.g., greater than about 10 amino acids), the peptide is commonly called a polypeptide or a protein. While the term "protein" encompasses the term "polypeptide", a "polypeptide" may be a less than full-length protein.[0080] As used herein a "LAMP polypeptide" refers to the mammalian lysosomal associated membrane proteins human LAMP-1, human LAMP-2, human LAMP-3, human LIMP-2, human Endolyn, human LIMBIC, human LAMP-5, or human Macrosailin as described herein, as well as orthologs (such as, for example, the LAMP proteins shown in Figures 3-10), and allelic variants.
WO 2018/204534 PCT/US2018/030725 id="p-81" id="p-81" id="p-81"
[0081] As used herein, "expression" refers to the process by which polynucleotides are transcribed into mRNA and/or translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA transcribed from the genomic DNA.[0082] As used herein, "under transcriptional control" or "operably linked" refers to expression (e.g., transcription or translation) of a polynucleotide sequence which is controlled by an appropriate juxtaposition of an expression control element and a coding sequence. In one aspect, a DNA sequence is "operatively linked" to an expression control sequence when the expression control sequence controls and regulates the transcription of that DNA sequence.[0083] As used herein, "coding sequence" is a sequence which is transcribed and translated into a polypeptide when placed under the control of appropriate expression control sequences. The boundaries of a coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxyl) terminus. A coding sequence can include, but is not limited to, a prokaryotic sequence, cDNA from eukaryotic mRNA, a genomic DNA sequence from eukaryotic (e.g., mammalian) DNA, and even synthetic DNA sequences. A polyadenylation signal and transcription termination sequence will usually be located 3' to the coding sequence.[0084] As used herein, two coding sequences "correspond" to each other if the sequences or their complementary sequences encode the same amino acid sequences.[0085] As used herein, "signal sequence" denotes the endoplasmic reticulum translocation sequence. This sequence encodes a signal peptide that communicates to a cell to direct a polypeptide to which it is linked (e.g., via a chemical bond) to an endoplasmic reticulum vesicular compartment, to enter an exocytic/endocytic organelle, to be delivered either to a cellular vesicular compartment, the cell surface or to secrete the polypeptide. This signal sequence is sometimes clipped off by the cell in the maturation of a protein. Signal sequences can be found associated with a variety of proteins native to prokaryotes and eukaryotes.[0086] As used herein, "trafficking" denotes movement or progression of the polypeptide encoded by the improved LAMP Construct through cellular organelles or compartments in the pathway from the rough endoplasmic reticulum to the endosomal/lysosomal compartment or related organelles where antigen processing and binding to MHC II occurs.[0087] As used herein, an "improved LAMP Construct" and an "improved LAMP Construct comprising an antigen" and an "improved LAMP Construct comprising an antigen of interest" are used interchangeably. The different arrangements of the improved LAMP Constructs are illustrated in Figure 1 as ILC1-ILC6. Moreover, the use of an "improved LAMP Construct" encompasses both the polynucleotide sequence of the improved LAMP Construct as well as the protein encoded by the polynucleotide sequence of the improved LAMP Construct.[0088] As used herein, an "improved LAMP Construct delivery vehicle" is defined as any molecule or group of molecules or macromolecules that can carry an improved LAMP Construct into a host cell (e.g., such WO 2018/204534 PCT/US2018/030725 as genes or gene fragments, antisense molecules, ribozymes, aptamers, and the like) and which occurs in association with an improved LAMP Construct as described herein.[0089] As used herein, "improved LAMP Construct delivery," or "improved LAMP Construct transfer," refers to the introduction of the improved LAMP Construct into a host cell, irrespective of the method used for the introduction. The introduced improved LAMP Constructs may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced improved LAMP Construct either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.[0090] As used herein, a "viral improved LAMP Construct" refers to a virus or viral particle that comprises the improved LAMP Construct to be delivered into a host cell, either in vivo, ex vivo or in vitro. Examples of viral improved LAMP Constructs include, but are not limited to, adenovirus vectors, adeno- associated virus vectors, retroviral vectors, and the like. In aspects where gene transfer is mediated by an adenoviral vector, an improved LAMP Construct includes the adenovirus genome or part thereof, and a selected, non-adenoviral gene, in association with adenoviral capsid proteins.[0091] As used herein, "adenoviral-mediated gene transfer" or "adenoviral transduction" refers to the process by which an improved LAMP Construct is transferred into a host cell by virtue of the adenovirus entering the cell. Preferably, the improved LAMP Construct is able to replicate and/or integrate and be transcribed within the cell.[0092] As used herein, "adenovirus particles" are individual adenovirus virions comprised of an external capsid and an improved LAMP Construct, where the capsid is further comprised of adenovirus envelope proteins. The adenovirus envelope proteins may be modified to comprise a fusion polypeptide which contains a polypeptide ligand covalently attached to the viral protein, e.g., for targeting the adenoviral particle to a particular cell and/or tissue type.[0093] As used herein, the term "administering" or "immunizing" or "injecting" an improved LAMP Construct refers to transducing, transfecting, microinjecting, electroporating, or shooting the cell with the improved LAMP Construct. In some aspects, improved LAMP Constructs are introduced into a target cell by contacting the target cell with a delivery cell (e.g., by cell fusion or by lysing the delivery cell when it is in proximity to the target cell).[0094] As used herein, the phrase "prime boost" describes the use of an improved LAMP Construct described herein used to prime a T-cell response followed by the use of a second improved LAMP Construct comprising an antigen, a DNA vaccine comprising an antigen or a recombinant antigen to boost the response (or visa versa). These heterologous prime-boost immunizations elicit immune responses of greater height and breadth than can be achieved by priming and boosting with the same vector. The priming with an improved LAMP Construct comprising an antigen initiates memory cells; the boost step expands the memory response. Preferably, two different agents that do not raise responses against each other are used and thus do not interfere WO 2018/204534 PCT/US2018/030725 with each other's activity. Mixtures of antigens are specifically contemplated in the prime and/or boost step. Boosting can occur once or multiple times.[0095] As used herein, "hybridization" refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence- specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. A hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.[0096] As used herein, a polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) which has a certain percentage (for example, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 99%) of "sequence identity" to another sequence means that, when maximally aligned, using software programs routine in the art, that percentage of bases (or amino acids) are the same in comparing the two sequences.[0097] Two sequences are "substantially homologous" or "substantially similar" when at least about 50%, at least about 60%, at least about 70%, at least about 75%, and preferably at least about 80%, and most preferably at least about 90 or 95% of the nucleotides match over the defined length of the DNA sequences. Similarly, two polypeptide sequences are "substantially homologous" or "substantially similar" when at least about 50%, at least about 60%, at least about 66%, at least about 70%, at least about 75%, and preferably at least about 80%, and most preferably at least about 90 or 95% of the amino acid residues of the polypeptide match over a defined length of the polypeptide sequence. Sequences that are substantially homologous can be identified by comparing the sequences using standard software available in sequence data banks. Substantially homologous nucleic acid sequences also can be identified in a Southern hybridization experiment under, for example, stringent conditions as defined for that particular system. Defining appropriate hybridization conditions is within the skill of the art. For example, stringent conditions can be: hybridization at 5xSSC and 50% formamide at 42°C, and washing at O.lxSSC and 0.1% sodium dodecyl sulfate at 60°C. Further examples of stringent hybridization conditions include: incubation temperatures of about 25 degrees C to about 37 degrees C; hybridization buffer concentrations of about 6xSSC to about WxSSC; formamide concentrations of about 0% to about 25%; and wash solutions of about 6xSSC. Examples of moderate hybridization conditions include: incubation temperatures of about 40 degrees C to about 50 degrees C.; buffer concentrations of about 9xSSC to about 2xSSC; formamide concentrations of about 30% to about 50%; and wash solutions of about 5xSSC to about 2xSSC. Examples of high stringency conditions include: incubation temperatures of about 55 degrees C to about 68 degrees C.; buffer concentrations of about IxSSC to about O.lxSSC; formamide concentrations of about 55% to about 75%; and wash solutions of about IxSSC, O.lxSSC, or deionized water. In general, hybridization incubation times are from 5 minutes to 24 hours, with 1, 2, or more washing steps, and wash WO 2018/204534 PCT/US2018/030725 incubation times are about 1, 2, or 15 minutes. SSC is 0.15 M NaCl and 15 mM citrate buffer. It is understood that equivalents of SSC using other buffer systems can be employed. Similarity can be verified by sequencing, but preferably, is also or alternatively, verified by function (e.g., ability to traffic to an endosomal compartment, and the like), using assays suitable for the particular domain in question.[0098] The terms "percent (%) sequence similarity", "percent (%) sequence identity", and the like, generally refer to the degree of identity or correspondence between different nucleotide sequences of nucleic acid molecules or amino acid sequences of polypeptides that may or may not share a common evolutionary origin (see Reeck et al., supra). Sequence identity can be determined using any of a number of publicly available sequence comparison algorithms, such as BLAST, PASTA, DNA Strider, GCG (Genetics Computer Group, Program Manual for the GCG Package, Version 7, Madison, Wisconsin), etc.[0099] To determine the percent identity between two amino acid sequences or two nucleic acid molecules, the sequences are aligned for optimal comparison purposes. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., percent identity = number of identical positions/total number of positions (e.g., overlapping positions) x 100). In one embodiment, the two sequences are, or are about, of the same length. The percent identity between two sequences can be determined using techniques similar to those described below, with or without allowing gaps. In calculating percent sequence identity, typically exact matches are counted.[0100] The determination of percent identity between two sequences can be accomplished using a mathematical algorithm. A non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, Proc. Natl. Acad. Sci. USA 1990, 87:2264, modified as in Karlin and Altschul, Proc. Natl. Acad. Sci. USA 1993, 90:5873-5877. Such an algorithm is incorporated into the NBLAST andXBLAST programs of Altschul et al, J. Mol. Biol. 1990; 215: 403. BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12, to obtain nucleotide sequences homologous to sequences of the invention. BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3, to obtain amino acid sequences homologous to protein sequences of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al, Nucleic Acids Res. 1997, 25:3389. Alternatively, PSI-Blast can be used to perform an iterated search that detects distant relationship between molecules. See Altschul et al. (1997) supra. When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See ncbi.nlm.nih.gov/BLAST/ on the WorldWideWeb.[0101] Another non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, CAB IOS 1988; 4: 1 1-17. Such an algorithm is incorporated into the ALIGN program (version 2.0), which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used.
WO 2018/204534 PCT/US2018/030725 id="p-102" id="p-102" id="p-102"
[0102] In a preferred embodiment, the percent identity between two amino acid sequences is determined using the algorithm of Needleman and Wunsch (J. Mol. Biol. 1970, 48:444-453), which has been incorporated into the GAP program in the GCG software package (Accelrys, Burlington, MA; available at accelrys.com on the WorldWideWeb), using either a Blossum 62 matrix or a PAM250 matrix, a gap weight of 16, 14, 12, 10, 8, 6, or 4, and a length weight of 1, 2, 3, 4, 5, or 6. In yet another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package using a NWSgapdna.CMP matrix, a gap weight of 40, 50, 60, 70, or 80, and a length weight of 1, 2, 3, 4, 5, or 6. A particularly preferred set of parameters (and the one that can be used if the practitioner is uncertain about what parameters should be applied to determine if a molecule is a sequence identity or homology limitation of the invention) is using a Blossum 62 scoring matrix with a gap open penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.[0103] Another non-limiting example of how percent identity can be determined is by using software programs such as those described in Current Protocols In Molecular Biology (F. M. Ausubel et al., eds., 1987) Supplement 30, section 7.7.18, Table 7.7.1. Preferably, default parameters are used for alignment. A preferred alignment program is BLAST, using default parameters. In particular, preferred programs are BLASTN and BLASTP, using the following default parameters: Genetic code=standard; filter=none; strand=both; cutoff=60; expect=10; Matrix=BLOSUM62; Descriptions=50 sequences; sort by=HIGH SCORE; Databases=non- redundant, GenBank+EMBL+DDBJ+PDB+GenBank CDS translations+SwissProtein+SPupdate+PIR. Details of these programs can be found at the following Internet address: http://www.ncbi.nlm.nih.gov/cgi-bin/BLAST.[0104] Statistical analysis of the properties described herein may be carried out by standard tests, for example, t-tests, ANOVA, or Chi squared tests. Typically, statistical significance will be measured to a level of p=0.05 (5%), more preferably p=0.01, p=0.001, p=0.0001, p=0.000001[0105] "Conservatively modified variants" of domain sequences also can be provided. With respect to particular nucleic acid sequences, conservatively modified variants refer to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Specifically, degenerate codon substitutions can be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer, et al., 1991, Nucleic Acid Res. 19: 5081; Ohtsuka, et al., 1985, J. Biol. Chern. 260: 2605-2608; Rossolini et al., 1994, Mol. Cell. Probes 8: 91-98).[0106] The term "biologically active fragment", "biologically active form", "biologically active equivalent" of and "functional derivative" of a wild-type protein, possesses a biological activity that is at least substantially equal (e.g., not significantly different from) the biological activity of the wild type protein as measured using an assay suitable for detecting the activity.[0107] As used herein, "in vivo" nucleic acid delivery, nucleic acid transfer, nucleic acid therapy" and the like, refer to the introduction of an improved LAMP Construct directly into the body of an organism, such WO 2018/204534 PCT/US2018/030725 as a human or non-human mammal, whereby the improved LAMP Construct is introduced to a cell of such organism in vivo.[0108] As used herein, the term "in situ" refers to a type of in vivo nucleic acid delivery in which the improved LAMP Construct is brought into proximity with a target cell (e.g., the nucleic acid is not administered systemically). For example, in situ delivery methods include, but are not limited to, injecting an improved LAMP Construct directly at a site (e.g., into a tissue, such as a tumor or heart muscle), contacting the improved LAMP Construct with cell(s) or tissue through an open surgical field, or delivering the improved LAMP Constructs to a site using a medical access device such as a catheter.[0109] As used herein, the term "isolated" or "purified" means separated (or substantially free) from constituents, cellular and otherwise, in which the polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, are normally associated with in nature. For example, with respect to an improved LAMP Construct, an isolated polynucleotide is one that is separated from the 5' and 3' sequences with which it is normally associated in the chromosome. As is apparent to those of skill in the art, a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, does not require "isolation" to distinguish it from its naturally occurring counterpart. By substantially free or substantially purified, it is meant at least 50% of the population, preferably at least 70%, more preferably at least 80%, and even more preferably at least 90%, are free of the components with which they are associated in nature.[0110] As used herein, a "target cell" or "recipient cell" refers to an individual cell or cell which is desired to be, or has been, a recipient of the improved LAMP Constructs described herein. The term is also intended to include progeny of a single cell, and the progeny may not necessarily be completely identical (in morphology or in genomic or total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. A target cell may be in contact with other cells (e.g., as in a tissue) or may be found circulating within the body of an organism.[0111] As used herein, a "subject" is a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets. In other preferred embodiments, the "subject" is a rodent (e.g. a rat, a mouse, a rabbit, a llama, camels, a cow, a guinea pig, a hamster, a dog, a cat, a horse, a non-human primate, a simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon, rhesus macaque), or an ape (e.g. gorilla, chimpanzee, orangutan, gibbon). In other embodiments, non-human mammals, especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.[0112] The terms "cancer," "neoplasm," and "tumor," are used interchangeably and in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism. Primary cancer cells transformation that makes them pathological to the host organism. Primary cancer cells (that is, cells obtained from near the site of malignant transformation) can be WO 2018/204534 PCT/US2018/030725 readily distinguished from non-cancerous cells by well-established techniques, particularly histological examination. The definition of a cancer cell, as used herein, includes not only a primary cancer cell, but any cell derived from a cancer cell ancestor. This includes metastasized cancer cells, and in vitro cultures and cell lines derived from cancer cells. When referring to a type of cancer that normally manifests as a solid tumor, a "clinically detectable" tumor is one that is detectable on the basis of tumor mass, e.g., by procedures such as CAT scan, MR imaging, X-ray, ultrasound or palpation, and/or which is detectable because of the expression of one or more cancer-specific antigens in a sample obtainable from a patient.[0113] In preferred embodiments, the cancer (including all stages of progression, includinghyperplasia) is an adenocarcinoma, sarcoma, skin cancer, melanoma, bladder cancer, brain cancer (including glioblastoma multiforme), breast cancer, uterine cancer, ovarian cancer, prostate cancer, lung cancer (including, but not limited to NSCLC, SCLC, squamous cell cancer), colorectal cancer, anal cancer, rectal cancer, cervical cancer, liver cancer, head and neck cancer, oral cancer, salivary gland cancer, esophageal cancer, pancreas cancer, pancreatic ductal adenocarcinoma (PDA), renal cancer, stomach cancer, kidney cancer, multiple myeloma or cerebral cancer. Also, preferred diseases to be treated with the LAMP Constructs described herein include those hyperproliferative disorders or cancer caused by cytomegalovirus (CMV).[0114] As used herein, the term "pharmaceutically acceptable carrier" encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. Compositions comprising the improved LAMP Constructs also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Martin Remington's Pharm. Sci., 15th Ed. (Mack Puhi. Co., Easton (1975)).[0115] A cell has been "transformed", "transduced", or "transfected" by the improved LAMP Constructs when such nucleic acids have been introduced inside the cell. Transforming DNA may or may not be integrated (covalently linked) with chromosomal DNA making up the genome of the cell. In prokaryotes, yeast, and mammalian cells for example, the improved LAMP Constructs may be maintained on an episomal element, such as a plasmid. In a eukaryotic cell, a stably transformed cell is one in which the improved LAMP Constructs have become integrated into a chromosome so that it is inherited by daughter cells through chromosome replication. This stability is demonstrated by the ability of the eukaryotic cell to establish cell lines or clones comprised of a population of daughter cells containing the improved LAMP Constructs. A "clone" is a population of cells derived from a single cell or common ancestor by mitosis. A "cell line" is a clone of a primary cell that is capable of stable growth in vitro for many generations (e.g., at least about 10).[0116] As used herein, an "effective amount" is an amount sufficient to affect beneficial or desired results, e.g., such as an effective amount of the improved LAMP Construct transfer and/or expression, and/or the attainment of a desired therapeutic endpoint. An effective amount can be administered in one or more administrations, applications or dosages. In one aspect, an effective amount of an improved LAMP Construct is WO 2018/204534 PCT/US2018/030725 an amount sufficient to transform/transduce/transfect at least one cell in a population of cells comprising at least two cells.[0117] As used herein, a "therapeutically effective amount" is used herein to mean an amount sufficient to prevent, correct and/or normalize an abnormal physiological response. In one aspect, a "therapeutically effective amount" is an amount sufficient to reduce by at least about 30 percent, more preferably by at least percent, most preferably by at least 90 percent, a clinically significant feature of pathology, such as for example, size of a tumor mass, antibody production, cytokine production, fever or white cell count, etc.[0118] An "antibody" is any immunoglobulin, including antibodies and fragments thereof, that binds a specific antigen. The term encompasses polyclonal, monoclonal, and chimeric antibodies (e.g., bispecific antibodies). An "antibody combining site" is that structural portion of an antibody molecule comprised of heavy and light chain variable and hypervariable regions that specifically binds antigen. Exemplary antibody molecules are intact immunoglobulin molecules, substantially intact immunoglobulin molecules, and those portions of an immunoglobulin molecule that contains the paratope, including Fab, Fab', F(ab')2 and F(v) portions, which portions are preferred for use in the therapeutic methods described herein. Thus, the term antibody encompasses not only whole antibody molecules, but also antibody fragments as well as variants (including derivatives such as fusion proteins) of antibodies and antibody fragments. Examples of molecules which are described by the term "antibody" in this application include, but are not limited to: single chain Fvs (scFvs), Fab fragments, Fab' fragments, F(ab')2, disulfide linked Fvs (sdFvs), Fvs, and fragments comprising or alternatively consisting of, either a VL or a VH domain. The term "single chain Fv" or "scFv" as used herein refers to a polypeptide comprising a VL domain of an antibody linked to a VH domain of an antibody. See Carter (2006) Nature Rev. Immunol. 6:243.[0119] Additionally, antibodies of the invention include, but are not limited to, monoclonal, multi- specific, bi-specific, human, humanized, mouse, or chimeric antibodies, single chain antibodies, camelid antibodies, Fab fragments, F(ab') fragments, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), domain antibodies and epitope-binding fragments of any of the above. The immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.[0120] Most preferably, the antibodies are human antibodies. As used herein, "human" antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries and xenomice or other organisms that have been genetically engineered to produce human antibodies. The improved LAMP Constructs described herein can be used in combination with known techniques for generating human antibodies and human monoclonal antibodies as described in the exemplified protocols, see, e.g., PCT publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0598877; U.S.Patent Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825; 5,661,016; WO 2018/204534 PCT/US2018/030725 ,545,806; 5,814,318; 5,885,793; 5,916,771; and 5,939,598; andLonberg andHuszar, Int. Rev. Immunol. 13:65- (1995).[0121] Human antibodies or "humanized" chimeric monoclonal antibodies can be produced using the improved LAMP Constructs in combination with techniques described herein or otherwise known in the art. For example, standard methods for producing chimeric antibodies are known in the art. See, for review the following references: Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Cabilly et al., U.S. Patent No. 4,816,567; Taniguchi et al., EP 171496; Morrison et al., EP 173494; Neuberger et al., WO 8601533; Robinson et al., WO 8702671; Boulianne et al., Nature 312:643 (1984); Neuberger et al., Nature 314:268 (1985).[0122] The antibodies of the present invention may be monovalent, bivalent, trivalent or multivalent. For example, monovalent scFvs can be multimerized either chemically or by association with another protein or substance. A scFv that is fused to a hexahistidine tag or a Flag tag can be multimerized using Ni-NTA agarose (Qiagen) or using anti-Flag antibodies (Stratagene, Inc.). Additionally, the improved LAMP Constructs can be used to generate monospecific, bispecific, trispecific or of greater multispecificity for the encoded antigen(s) contained in the improved LAMP Construct. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S. Patent Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; 5,601,819; Kostelny et.al., J. Immunol. 148:1547-1553 (1992).[0123] An "epitope" is a structure, usually made up of a short peptide sequence or oligosaccharide, that is specifically recognized or specifically bound by a component of the immune system. T-cell epitopes have generally been shown to be linear oligopeptides. Two epitopes correspond to each other if they can be specifically bound by the same antibody. Two epitopes correspond to each other if both are capable of binding to the same B cell receptor or to the same T cell receptor, and binding of one antibody to its epitope substantially prevents binding by the other epitope (e.g., less than about 30%, preferably, less than about 20%, and more preferably, less than about 10%, 5%, 1%, or about 0.1% of the other epitope binds). In the present invention, multiple epitopes can make up an antigen.[0124] The term "antigen" or "antigen of interest" as used herein covers any polypeptide sequence encoded by a polynucleotide sequence cloned into the improved LAMP Construct which is used to elicit an innate or adaptive immune response as shown in Table 1. An "antigen" encompasses both a single antigen as well as multiple antigenic sequences (derived from the same or different proteins) cloned into the improved LAMP Construct.[0125] The term "antigen presenting cell" as used herein includes any cell which presents on its surface an antigen in association with a major histocompatibility complex molecule, or portion thereof, or, alternatively, one or more non-classical MHC molecules, or a portion thereof. Examples of suitable APCs are discussed in detail below and include, but are not limited to, whole cells such as macrophages, dendritic cells, B cells, hybrid APCs, and foster antigen presenting cells.
WO 2018/204534 PCT/US2018/030725 id="p-126" id="p-126" id="p-126"
[0126] As used herein an "engineered antigen-presenting cell" refers to an antigen-presenting cell that has a non-natural molecular moiety on its surface. For example, such a cell may not naturally have a costimulator on its surface or may have an additional artificial costimulator in addition to a natural costimulator on its surface, or may express a non-natural class II molecule on its surface. In preferred embodiments, the engineered antigen- presenting cell has the antigen expressed from the improved LAMP Construct on its surface.[0127] As used herein, "immune effector cells" refers to cells capable of binding an antigen and which mediate an immune response. These cells include, but are not limited to, T cells, B cells, monocytes, macrophages, NK cells and cytotoxic T lymphocytes (CTLs), for example CTL lines, CTL clones, and CTLs from tumor, inflammatory, or other infiltrates.[0128] As used herein, "partially human" refers to a nucleic acid having sequences from both a human and a non-human vertebrate. In the context of partially human sequences, the partially human nucleic acids have sequences of human immunoglobulin coding regions and sequences based on the non-coding sequences of the endogenous immunoglobulin region of the non-human vertebrate. The term "based on" when used with reference to endogenous non-coding sequences from a non-human vertebrate refers to sequences that correspond to the non-coding sequence and share a relatively high degree of homology with the non-coding sequences of the endogenous loci of the host vertebrate, e.g., the non-human vertebrate from which the ES cell is derived. Preferably, the non-coding sequences share at least an 80%, more preferably 90% homology with the corresponding non-coding sequences found in the endogenous loci of the non-human vertebrate host cell into which a partially human molecule comprising the non-coding sequences has been introduced.[0129] The term "immunoglobulin variable region" as used herein refers to a nucleotide sequence that encodes all or a portion of a variable region of an antibody molecule or all or a portion of a regulatory nucleotide sequence that controls expression of an antibody molecule. Immunoglobulin regions for heavy chains may include but are not limited to all or a portion of the V, D, J, and switch regions, including introns. Immunoglobulin region for light chains may include but are not limited to the V and J regions, their upstream flanking sequences, introns, associated with or adjacent to the light chain constant region gene.[0130] By "transgenic animal" is meant a non-human animal, usually a mammal, having an exogenous nucleic acid sequence present as an extrachromosomal element in a portion of its cells or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells). In generating a transgenic animal comprising human sequences, a partially human nucleic acid is introduced into the germ line of such transgenic animals by genetic manipulation of, for example, embryos or embryonic stem cells of the host animal according to methods well known in the art.[0131] A "vector" includes plasmids and viruses and any DNA or RNA molecule, whether self- replicating or not, which can be used to transform or transfect a cell.[0132] An "isolated" or "purified" population of cells is substantially free of cells and materials with which it is associated in nature. By substantially free or substantially purified APCs it is meant at least 50% of WO 2018/204534 PCT/US2018/030725 the population are APCs, preferably at least 70%, more preferably at least 80%, and even more preferably at least 90%, are free of non-APCs cells with which they are associated in nature.[0133] As used herein, a "genetic modification" refers to any addition, deletion or disruption to a cell's normal nucleotides. Any method which can achieve the genetic modification of APCs are within the spirit and scope of this invention. Art recognized methods include viral mediated gene transfer, liposome mediated transfer, transformation, transfection and transduction, e.g., viral-mediated gene transfer such as the use of the improved LAMP Constructs based on DNA viruses such as adenovirus, adeno-associated virus and herpes virus, as well as retroviral based vectors.[0134] The practice of the present invention employs, unless otherwise indicated, conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Maniatis, Fritsch & Sambrook, In Molecular Cloning: A Laboratory Manual (1982); DNA Cloning: A Practical Approach, Volumes I and II (D. N. Glover, ed., 1985); Oligonucleotide Synthesis (M. J. Gait, ed., 1984); Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins, eds., 1985); Transcription and Translation (B. D. Hames & S. I. Higgins, eds., 1984); Animal Cell Culture (R. I. Freshney, ed., 1986); Immobilized Cells and Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide to Molecular Cloning (1984).[0135] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing devices, formulations and methodologies that may be used in connection with the presently described invention LAMP Constructs [0136] LAMP-1, as deduced from a cDNA clone (Chen, et al., J. Biol. Chern. 263: 8754, 1988) consists of a polypeptide core of about 382 amino acids with a large (346-residue) lumenal amino-terminal domain followed by a 24-residue hydrophobic transmembrane region and short (12-residue) carboxyl-terminal cytoplasmic tail. See, Figure 2A and 2B. The lumenal domain is highly glycosylated, being substituted with about 20 asparagine linked complex-type oligosaccharides and consists of two approximately 160-residue "homology domains" that are separated by a proline/serine-rich hinge region. Each of these "homology domains" contains 4 uniformly spaced cysteine residues, disulfide bonded to form four 36-38-residue loops symmetrically placed within the two halves of the lumenal domain (Arterburn, et al., J. Biol. Chern. 265: 7419, 1990; see, also Chen, et al., J. Biol. Chern. 25: 263(18): 8754-8, 1988). Figure 2A schematically shows the conserved domains between LAMP-1, LAMP-2, LAMP-3, Endolyn, LIMBIC, LAMP5, or Macrosailin.[0137] Previously reported LAMP constructs comprise the following elements in this specific arrangement: (a) a full lumenal domain of LAMP-1 protein, the antigen and then the full transmembrane/cytoplasmic tail of LAMP-1 protein; or (b) the antigen and the full transmembrane/cytoplasmic WO 2018/204534 PCT/US2018/030725 tail of a LAMP-1 protein. In example (a), the antigenic sequence is inserted in between the full lumenal domain of a LAMP-1 protein and the LAMP-1 full transmembrane domain/cytoplasmic tail. Both constructs have been shown to successfully target an antigenic sequence to the lysosome/endosome and will be referred to as "complete LAMP Constructs" as shown in Figure 1 as compared to the improved LAMP Constructs ILC1-ILCdescribed herein. The improved LAMP Constructs described herein do not include the complete LAMP Constructs described in the prior art.[0138] Although it has been widely reported in the literature that fragments smaller than the full lumenal domain of LAMP-1 were not effective in generating a robust immune response (see, e.g. Godinho et al.). In contrast, the inventors unexpectedly discovered that specific fragments, in certain arrangements, did in fact effectively present antigens to the immune system, generating in many cases a more robust immune response, including the generation of a different repertoire of antibodies. For example, the inventors have identified that the minimal LAMP lumenal domain fragment that is effective for generating a robust immune response is not the full lumenal domain (as widely reported in the literature) but rather a single Homology Domain of the Lumenal Domain of a LAMP Protein.[0139] For example, constructs can comprise, not the full lumenal domain, but instead a single Homology Domain of the Lumenal Domain of a LAMP Protein. As used herein, the "Homology Domain" comprises at least the 4 uniformly spaced cysteine residues shown in Figures 3-10. These cysteine resides are labeled 1, 2, 3, and 4 (and in LIMP-2 and Macrosailin - five cysteines are identified, LIMBIC - six cysteines are identified and Endolyn - eight cysteines are identified) in each Homology Domain as shown in Figures 3- and are defined herein as the "Cysteine Conserved Fragment." Additional amino acids can be included to either the N-terminus end and/or the C-terminus end of the Cysteine Conserved Fragment to generate, up to and including a full Homology Domain of a LAMP protein. These additional added amino acids can be derived from the Homology Domain from which the Cysteine Conserved Fragment is derived or from other LAMP Protein Homology Domains. Thus, as used herein, a LAMP Homology Domain comprises and/or consists of one Cysteine Conserved Fragment. At least two LAMP Homology Domains make up the Lumenal Domain of LAMP-1, LAMP-2, LAMP-3, or Endolyn.[0140] Specifically, in one preferred embodiment, the improved LAMP Construct comprises at least one cancer antigen described in Table 1 (preferably the sequences/fragments/epitopes described in column 4 of Table 1) and/or the combinations described in paragraph [150] fused to the N-terminus of the lumenal domain of a LAMP protein, at least one homology domain of a LAMP protein, or at least one Cysteine Conserved Fragment of a LAMP protein. See, for example ILC-2 and ILC-6 of Figure 1. In preferred embodiments, these constructs also comprise a transmembrane domain of a LAMP protein, and/or the cytosolic tail of a LAMP protein. In other preferred embodiments, when an antigen contains a transmembrane domain, the transmembrane domain of a LAMP protein and/or the cytosolic tail of a LAMP protein is unnecessary. In preferred embodiments, two homology domains are included in the improved LAMP Construct (e.g., ILC-1 of WO 2018/204534 PCT/US2018/030725 Figure 1). In further preferred embodiments, the two homology domains are derived from a LAMP-1, LAMP- 2, LAMP-3, or an Endolyn protein. Alternatively, the two homology domains are derived from different LAMP proteins. In these constructs comprising two homology domains, a LAMP hinge domain may also be included. The improved LAMP Constructs described in this paragraph are unexpected in view of the prior art as the antigen has always been placed in between the full lumenal LAMP-1 domain and the full LAMP-transmembrane/cytoplasmic tail, as fragments of the lumenal domain have not been reported to be effective in generating a robust immume response.[0141] In another preferred embodiment, the improved LAMP Construct comprises at least one cancer antigen described in Table 1 (preferably the sequences/fragments/epitopes described in column 4 of Table 1) and/or the combinations described in paragraph [150] fused to the C-terminus of a single homology domain of a LAMP protein or a single Cysteine Conserved Fragment of a LAMP protein (e.g., ILC-5 of Figure 1). In preferred embodiments, these constructs also comprise a transmembrane domain of a LAMP protein, and/or the cytosolic tail of a LAMP protein (e.g., ILC-3 of Figure 1) In other preferred embodiments, when an antigen contains a transmembrane domain, the transmembrane domain of a LAMP protein and/or the cytosolic tail of a LAMP protein is unnecessary. Cancer antigens described in Table 1 can also be inserted into the complete LAMP constructs as depicted in Figure 1. Alternatively, two homology domains from two different LAMP proteins may be used. The improved LAMP Constructs described in this paragraph are unexpected in view of the prior art as the antigen has always been placed in between the full lumenal LAMP-1 domain and the full LAMP-1 transmembrane/cytoplasmic tail, as fragments of the lumenal domain have not been reported to be effective in generating a robust immune response.[0142] Thus, the improved LAMP Construct comprises at least one cancer antigen described in Table (preferably the sequences/fragments/epitopes described in column 4 of Table 1) and/or the combinations described in paragraph [150] fused to the C-terminus of a single homology domain of a LAMP protein or a single Cysteine Conserved Fragment of a LAMP protein. See, for example, ILC-3 and ILC-5 of Figure 1. In preferred embodiments, these constructs also comprise a transmembrane domain of a LAMP protein, and/or the cytosolic tail of a LAMP protein. In other preferred embodiments, when an antigen contains a transmembrane domain, the transmembrane domain of a LAMP protein and/or the cytosolic tail of a LAMP protein is unnecessary. The improved LAMP Constructs described in this paragraph are unexpected in view of the prior art as described above.[0143] In another preferred embodiment, the improved LAMP Construct comprises at least one antigen of interest fused in between a first homology domain of a LAMP protein and a second homology domain of a LAMP protein (or at least between two Cysteine Conserved Fragments). See, for example, ILC-4 of Figure 1. In preferred embodiments, the two homology domains are derived from LAMP-1, LAMP-2, LAMP-3, or an Endolyn protein. In these constructs, the antigen may be placed in the LAMP hinge region. Alternatively, two homology domains from two different LAMP proteins may be used. This arrangement of at least one cancer WO 2018/204534 PCT/US2018/030725 antigen described in Table 1 (preferably the sequences/fragments/epitopes described in column 4 of Table 1) and/or the combinations described in paragraph [150] fused in between two LAMP homology domains (including Cysteine Conserved Fragments) is unexpected in view of the prior art as described above.[0144] Each of the improved LAMP Constructs defined above can be generated using the domains defined in the Figures. For example, it is specifically contemplated that the domains included in the improved LAMP Construct illustrated in Figure 1, for example, can originate from sequences derived from orthologous sequences. See, Figures 3-10 for example. It is expressly contemplated that the equivalent domains defined in Figures 2A and 2B be used to generate the improved LAMP Constructs illustrated in Figure 1 for orthologous sequences. Moreover, the orthologous sequences shown in Figures 3-10 are representative of the sequences that can be used to generate the domains. It is well within the skill in the art to identify other orthologous sequences and/or isotypes and comparing them to the alignments shown in Figures 3-10. Thus, by identifying the equivalent boundaries defined in Figure 2A and 2B for a human LAMP protein with the alignments shown in Figures 3-10, one can generate the improved LAMP Constructs illustrated in Figure 1.[0145] As would be well understood by the skilled artisan, the boundaries of each domain are an approximation and may be adjusted at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids based on cloning considerations and restriction enzyme placement. Therefore, when a particular domain (e.g., a LAMP Homology Domain) is included in the improved LAMP Construct, the amino acids beginning and ending of the domain may be adjust by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids as those boundaries defined in Figure 2A.[0146] Each of the improved LAMP Constructs described above can additionally comprise a signal sequence and/or additional amino acids in between each domain for cloning purposes as is well known in the art. Additionally, the LAMP homologous domains, the LAMP lumenal domain, the LAMP transmembrane domain, and/or the LAMP cytosolic tail domain can originate from the same LAMP protein (e.g., human LAMP- 1) or different LAMP proteins (e.g., lumenal domain from human LAMP-1 and transmembrane domain from human LAMP-2, and/or mixing of orthologous domains in the same gene family (e.g., LAMP-1) or different gene family (LAMP-1 and LAMP-2).[0147] Polypeptide variants of the described LAMP Constructs are contemplated. For example, polypeptides at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, 96%, 97%, 98% or 99% identity to any of the improved LAMP Constructs described herein as well as polynucleotides encoding these variants. Variants of the improved LAMP Constructs retain the ability to function by targeting the antigenic sequence to the lysosome. For example, a modified lumenal sequence must retain the ability to traffic both membrane and non-membrane antigenic materials to an endosomal compartment with at least about 50%, at least about 60%, at least 70%, at least about 80%, at least about 90%, or at least about 100% efficacy compared to the original domain sequence, i.e., an efficacy that results in sufficient antigen presentation by a cell comprising the chimeric sequence for it to mount an immune WO 2018/204534 PCT/US2018/030725 response. In one aspect, sequences containing a suitable trafficking signal may be identified by constructing an improved LAMP Construct containing the well-characterized antigenic domain of ovalbumin, a transmembrane domain, and the cytoplasmic domain of a protein containing a putative lysosomal/endosomal targeting signal. Efficiency of targeting can be measured by determining the ability of antigen presenting cells, expressing the improved LAMP Construct, to stimulate HA epitope-specific, MHC class II restricted T-cells (see, e.g., Example of U.S. Pat. No. 5,633,234).[0148] Polynucleotides encoding any of the described improved LAMP Constructs are preferred embodiments of the invention, along with polynucleotides at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, 96%, 97%, 98% or 99% identity to any of the improved LAMP Construct polynucleotides described herein. Variants of the improved LAMP Constructs retain the ability to function by targeting the antigenic sequence to the lysosome. For example, a modified lumenal sequence must retain the ability to traffic both membrane and non-membrane antigenic materials to an endosomal compartment with at least about 50%, at least about 60%, at least 70%, at least about 80%, at least about 90%, or at least about 100% efficacy compared to the original domain sequence, i.e., an efficacy that results in sufficient antigen presentation by a cell comprising the chimeric sequence for it to mount an immune response. In one aspect, sequences containing a suitable trafficking signal may be identified by constructing an improved LAMP Construct containing the well-characterized antigenic domain of ovalbumin, a transmembrane domain, and the cytoplasmic domain of a protein containing a putative lysosomal/endosomal targeting signal. Efficiency of targeting can be measured by determining the ability of antigen presenting cells, expressing the improved LAMP Construct, to stimulate HA epitope-specific, MHC class II restricted T-cells (see, e.g., Example 5 of U.S. Pat. No. 5,633,234).
Cancer Antigens [0149] The following antigens shown in Table 1 can be cloned into each of the LAMP Constructs described herein using techniques well known to the skilled artisan. The sequences/fragments/epitopes described in the fourth column can be also cloned into the improved LAMP Constructs as described herein. Moreover, it is specifically contemplated that any one of the cancer antigen listed in Table 1 can be combined with any other antigen listed in Table 1 (including the sequences/fragments/epitopes described in the fourth column) and inserted into the improved LAMP Constructs as described herein.
SEQID NO: Protein Name of each Antigen Accession/ Derived From Preferred Epitopes/Fragments that can be cloned into the LAMP Constructs described herein, comprise at least.... 114 pp65 ABQ23593 The highlighted epitopes shown in Figure 20, which include LLQTGIHVRVSQPSL and/or ALPLKMLNIPSINVH and/or DQYVKVYLESFCEDV and/or IIKPGKISHIMLDVAFTSH and/or PQYSEHPTFTSQYRIQGKL and/or PPWQAGILARNLVPMV and/or KYQEFFWDANDIYRIFA wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 115 and SEQ ID NO: 116. 117 and118gB P06473 The highlighted epitopes shown in Figure 20, which include TTSAQTRSVYSQHVT and/or QLIPDDYSNTHSTRYV and/or VSVFETSGGLVVFWQ and/or NSAYEYVDYLFKRMIDLS wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 119 and SEQ ID NO: 120. 121 IE1 Pl3202 The highlighted epitopes shown in Figure 20, which include VLAELVKQIKVRVDMVRHRIKEHMLKKYTQ and/orIVPEDKREMWMACIKELH and/or KDELRRKMMYMCYRNIEFFTKNSAFPKTT and/or SVMKRRIEEICMKVFAQYI and/or AIAEESDEEEAIVAY and/or VKSEPVSEIEEVAPEEEEDG wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 122 and SEQ ID NO: 123.124Morphological transforming region II ("MTRII")AAA66543 SEQ ID NO:125126 US28 AMJ53524 SEQ ID NO:127 128 IGFBP2 NP_000588 (a) about amino acids 39-328 of SEQ ID NO: 128; (b) about amino acids 40-328 of SEQ ID NO:128; (c) about amino acids 43-135 of SEQ ID NO:128; (d) about amino acids 1-163 of SEQ ID NO: 128; (e) about amino acids 2-163 of SEQ ID NO: 128; (f) about amino acids 39-163 of SEQ ID NO: 128; (g) about amino acids 40-163 of SEQ ID NO: 128; (h) about amino acids 229-309 of SEQ ID NO: 128; (i) amino acids 2-3of SEQ ID NO: 128; and/or (!) about amino acids 1-328 of SEQ ID NO: 128129HCMVA Viral interleukin-("IL10")P17150 SEQID NO: 130 W O 2018/204534 PCT/US2018/030725 131HCMVM Membrane glycoprotein ULI("UL144")F5HAM0 SEQID NO: 132 133HCMVM Protein UL141("UL141")Q6RJQ3 SEQID NO: 134 135HCMVA Unique short USglycoprotein ("US 11")P09727 SEQID NO: 136 137 and138HCMV Envelope glycoprotein H OS ("HOS")A0A0G2TM81 The highlighted epitopes shown in Figure 20, which include TYNSSLRNSTVVRENAISFNFFQSYNQYYVFHMPR and/orDLTETLERYQQRLNTYALVSKDLASYRSFS and/orSHTTSGLHRPHFNQTCILFD and/or QLNRHSYLKDPDFLDAALDF wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 139 and SEQ ID NO: 140. 141HCMVT Viral transcription factor IE("IE2")Q6SWP7 The highlighted epitopes shown in Figure 20, which include RRGRVKIDEVSRMFR and/or GIQIIYTRNHEVKSE and/or LSTPFLMEHTMPVTHPPEVA wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 142 and SEQ ID NO: 143.144TERT Isoform ("TERT")NP_937983.2SEQID NO: 145SEQID NO: 146 147 Survivin NP_001125727 SEQ ID NO:147; representative constructs include SEQ ID NO:193-197. 148 Tetanus 1AF9_A The highlighted epitopes shown in Figure 20, which include PGINGKAIHLVNNESSE and/or FNNFTVSFWLRVPKVSASHLEQYGT and/orYVSIDKFRIFCKALNPKEIEKLYTSYLS and/orILRVGYNAPGIPLYKKMEAVKLRDLK wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 1and SEQ ID NO: 150. 151Cancer Testis Antigen NY-ESO-1 ("NY-ESO-1")NP-001318.1(SEQID NO: 151)Amino acids 1-155 of SEQ ID NO: 151Amino acids 80-155 of SEQ ID NO: 151 W O 2018/204534 PCT/US2018/030725 152 HER2AAA75493.1(SEQID NO: 152) Construct comprising at least one of: amino acids 23-652 and/or 690-1255 of SEQ ID NO: 152 wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 153 and SEQ ID NO: 154. 155 HER3NP_001973.(SEQ ID NO: 155) Construct comprising at least one of: amino acids 20-643, 665-1201, and/or 1209-13of SEQ ID NO: 155, wherein each epitope can be combined in any order and/or in any combination and cloned into a LAMP Construct described herein, wherein each epitope is preferably separated by a linker, such as, for example GPGPG or PMGLP. Representative construct inserts are shown as SEQ ID NO: 156 and SEQ ID NO: 157 158 HVEMNP_003811.2(SEQ ID NO: 158)Amino acids 39-202 (SEQ ID NO: 159) 160-163 HPV Constructs Sequences used to create the constructs: (a) HPV 16 E6 (SEQ ID NO: 160); (b) HPV E6 (SEQ ID NO:161); (c) HPV 16 E7 (SEQ ID NO:162); (d) HPV 18 E7 (SEQ ID NO: 163) Representative Constructs:SEQ ID NO: 164 (Representative HPV 16 E6-E7 Construct)SEQ ID NO: 165 (Representative HPV 18 E6-E7 Construct)SEQ ID NO: 166 (HPV 16 E6 - linker-HPV 18 E6-linker-HPV 16 E7-linker-HPV E7) 169-172 EBV ConstructsAny of the following sequences, in any combination, could be used to create the constructs (a) EBV EBNA1 (SEQ ID NO: 169); (b) EBV Truncated EBNA-1 (SEQ ID NO:170); (c) EBV gp350 (SEQ ID NO:171); (d) EBV LMP2 (SEQ ID NO:172). For example, a representative construct could comprise truncated EBNA-1 and LMP2. 174-175 HBV ConstructsAny of the following sequences could be used (in either orientation) to create the constructs:HBV Middle S Protein (SEQ ID NO: 174) and HBV X Protein (SEQ ID NO: 175).For example, a representative construct could comprise Middle S Protein - X protein.176 TIGIT SEQID NO: 176177 TEM8 SEQID NO: 177178 TEM1 SEQID NO: 178179 HER2ECD+TM SEQID NO: 179180 CEA SEQID NO: 180181 TARP SEQID NO: 181182 PROSTEIN SEQID NO: 182 W O 2018/204534 PCT/US2018/030725 183 PSMA SEQID NO: 183184 BIRC4 SEQID NO: 184185 Mucin-1 SEQID NO: 185186 Mucin-1 Isoform SEQID NO: 186187 CD40 Ligand SEQID NO: 187188 WT-1 SEQID NO: 188189 WT-1 Truncated SEQID NO: 189190 FRAME SEQID NO: 190191 LAGE-1 SEQID NO: 191192 MAGE A3 SEQID NO: 192198 lA01_HLA-A/m UniProtKB: P30443 SEQID NO: 198199 1A02 UniProtKB: P01892 SEQID NO: 199200 5T4 UniProtKB: Q13641 SEQ ID NO:200201 ACRBP UniProtKB: Q8NEB7 SEQ ID NO:201202 AFP UniProtKB: P02771 SEQ ID NO:202203 AKAP4 UniProtKB: Q5JQC9 SEQ ID NO:203204 alpha-actinin-_4/m UniProtKB: B4DSX0 SEQ ID NO:204205 alpha-actinin-_4/m UniProtKB: B4E337 SEQ ID NO:205206 alpha-actinin-_4/m UniProtKB: 043707 SEQ ID NO:206207alpha-methylacyl- coenzyme_A_racemaseUniProtKB: A0A024RE16 SEQ ID NO:207 208alpha-methylacyl- coenzyme_A_racemaseUniProtKB: A8KAC3 SEQ ID NO:208209 ANDR UniProtKB: P10275 SEQ ID NO:209210 ART-4 UniProtKB: Q9ULX3 SEQ ID NO:210211 ARTCl/m UniProtKB: P52961 SEQID NO:211212 AURKB UniProtKB: Q96GD4 SEQ ID NO:212213 B2MG UniProtKB: P61769 SEQID NO:213214 B3GN5 UniProtKB: Q9BYG0 SEQ ID NO:214215 B4GN1 UniProtKB: Q00973 SEQ ID NO:215216 B7H4 UniProtKB: Q7Z7D3 SEQ ID NO:216217 BAGE-1 UniProtKB: QI3072 SEQ ID NO:217218 BASI UniProtKB: P35613 SEQID NO:218219 BCL-2 UniProtKB: A9QXG9 SEQ ID NO:219220 bcr/abl UniProtKB: A9UEZ4 SEQ ID NO:220221 bcr/abl UniProtKB: A9UEZ7 SEQ ID NO:221222 bcr/abl UniProtKB: A9UEZ8 SEQ ID NO:222223 bcr/abl UniProtKB: A9UEZ9 SEQ ID NO:223224 bcr/abl UniProtKB: A9UF00 SEQ ID NO:224 W O 2018/204534 PCT/US2018/030725 225 bcr/abl UniProtKB: A9UF01 SEQ ID NO:225226 bcr/abl UniProtKB: A9UF03 SEQ ID NO:226227 bcr/abl UniProtKB: A9UF04 SEQ ID NO:227228 bcr/abl UniProtKB: A9UF05 SEQ ID NO:228229 bcr/abl UniProtKB: A9UF06 SEQ ID NO:229230 bcr/abl UniProtKB: A9UF08 SEQ ID NO:230231 beta-catenin/m UniProtKB: P35222 SEQ ID NO:231232 beta-catenin/m UniProtKB: Q8WYA6 SEQ ID NO:232233 BING-4 UniProtKB: 015213 SEQ ID NO:233234 BIRC7 UniProtKB: Q96CA5 SEQ ID NO:234235 BRCAl/m UniProtKB: A0A024R1V0 SEQ ID NO:235236 BRCAl/m UniProtKB: A0A024R1V7 SEQ ID NO:236237 BRCAl/m UniProtKB: A0A024R1Z8 SEQ ID NO:237238 BRCAl/m UniProtKB: A0A068BFX7 SEQ ID NO:238239 BRCAl/m UniProtKB: C6YB45 SEQ ID NO:239240 BRCAl/m UniProtKB: C6YB47 SEQ ID NO:240241 BRCAl/m UniProtKB: G3XAC3 SEQ ID NO:241242 BY55 UniProtKB: 095971 SEQ ID NO:242243 CAMEL UniProtKB: 095987 SEQ ID NO:243244 CASPA UniProtKB: Q92851-4 SEQ ID NO:244245 cathepsin_B UniProtKB: A0A024R374 SEQ ID NO:245246 cathepsin_B UniProtKB: P07858 SEQ ID NO:246247 cathepsin_L UniProtKB: A0A024R276 SEQ ID NO:247248 cathepsin_L UniProtKB: P07711 SEQ ID NO:248249 cathepsin_L UniProtKB: Q9HBQ7 SEQ ID NO:249250 CD1A UniProtKB: P06126 SEQ ID NO:250251 CD1B UniProtKB: P29016 SEQ ID NO:251252 CD1C UniProtKB: P29017 SEQ ID NO:252253 CD1D UniProtKB: P15813 SEQ ID NO:253254 CD1E UniProtKB: P15812 SEQ ID NO:254255 CD20 UniProtKB: Pl 1836 SEQ ID NO:255256 CD22 UniProtKB: 060926 SEQ ID NO:256257 CD22 UniProtKB: P20273 SEQ ID NO:257258 CD22 UniProtKB: Q0EAF5 SEQ ID NO:258259 CD276 UniProtKB: Q5ZPR3 SEQ ID NO:259260 CD33 UniProtKB: B4DF51 SEQ ID NO:260261 CD33 UniProtKB: P20138 SEQ ID NO:261262 CD33 UniProtKB: Q546G0 SEQ ID NO:262 W O 2018/204534 PCT/US2018/030725 263 CD3E UniProtKB: P07766 SEQ ID NO:263264 CD3Z UniProtKB: P20963 SEQ ID NO:264265 CD44_Isoform_ 1 UniProtKB: P16070 SEQ ID NO:265266 CD44_Isoform_6 UniProtKB: Pl6070-6 SEQ ID NO:266267 CD4 UniProtKB: P01730 SEQ ID NO:267268 CD52 UniProtKB: P31358 SEQ ID NO:268269 CD52 UniProtKB: Q6IBD0 SEQ ID NO:269270 CD52 UniProtKB: V9HWN9 SEQ ID NO:270271 CD55 UniProtKB: Bl API 5 SEQ ID NO:271272 CD55 UniProtKB: D3DT85 SEQ ID NO:272273 CD55 UniProtKB: D3DT86 SEQ ID NO:273274 CD55 UniProtKB: P08174 SEQ ID NO:274275 CD56 UniProtKB: Pl3591 SEQ ID NO:275276 CD80 UniProtKB: A0N0P2 SEQ ID NO:276277 CD80 UniProtKB: P33681 SEQ ID NO:277278 CD86 UniProtKB: P42081 SEQ ID NO:278279 CD8A UniProtKB: P01732 SEQ ID NO:279280 CDC27/m UniProtKB: G5EA36 SEQ ID NO:280281 CDC27/m UniProtKB: P30260 SEQ ID NO:281282 CDE30 UniProtKB: P28908 SEQ ID NO:282283 CDK4/m UniProtKB: A0A024RBB6 SEQ ID NO:283284 CDK4/m UniProtKB: Pl 1802 SEQ ID NO:284285 CDK4/m UniProtKB: Q6LC83 SEQ ID NO:285286 CDK4/m UniProtKB: Q96BE9 SEQ ID NO:286287 CDKN2A/m UniProtKB: D1LYX3 SEQ ID NO:287288 CDKN2A/m UniProtKB: G3XAG3 SEQ ID NO:288289 CDKN2A/m UniProtKB: K7PML8 SEQ ID NO:289290 CDKN2A/m UniProtKB: L8E941 SEQ ID NO:290291 CDKN2A/m UniProtKB: Q8N726 SEQ ID NO:291292 CEA RefSeq: NP_004354 SEQ ID NO:292293 CEAM6 UniProtKB: P40199 SEQ ID NO:293294 CH3L2 UniProtKB: QI5782 SEQ ID NO:294295 CLCA2 UniProtKB: Q9UQC9 SEQ ID NO:295296 CML28 UniProtKB: Q9NQT4 SEQ ID NO:296297 CML66 UniProtKB: Q96RS6 SEQ ID NO:297298 COA-l/m UniProtKB: Q5T124 SEQ ID NO:298299 coactosin-like_protein UniProtKB: Q14019 SEQ ID NO:299300 collagen_XXIII UniProtKB: L8EAS4 SEQ ID NO:300 W O 2018/204534 PCT/US2018/030725 301 collagen_XXIII UniProtKB: Q86Y22 SEQ ID NOG01302 COX-2 UniProtKB: Q6ZYK7 SEQ ID NO:302303 CP1B1 UniProtKB: QI6678 SEQ ID NO:303304 CSAG2 UniProtKB: Q9Y5P2-2 SEQ ID NO:304305 CSAG2 UniProtKB: Q9Y5P2 SEQ ID NO:305306 CT45A1 UniProtKB: Q5HYN5 SEQ ID NO:306307 CT55 UniProtKB: Q8WUE5 SEQ ID NO: 307308 CT-_9/BRD6 UniProtKB: Q58F21 SEQ ID NO:308309 CT AG2_Isoform_LAGE-1A UniProtKB: 075638-2 SEQ ID NO:309310 CTAG2_Isoform_LAGE-1B UniProtKB: 075638 SEQ ID NOG 10311 CTCFL UniProtKB: Q8NI51 SEQ ID NOG 11312 Cten UniProtKB: Q8IZW8 SEQ ID NOG 12313 cyclin_B 1 UniProtKB: P14635 SEQ ID NOG 13314 cyclin_D 1 UniProtKB: P24385 SEQ ID NOG 14315 cyp-B UniProtKB: P23284 SEQ ID NOG 15316 DAM-10 UniProtKB: P43366 SEQ ID NOG 16317 DEPIA UniProtKB: Q5TB30 SEQ ID NOG 17318 E7 UniProtKB: P03129 SEQ ID NOG 18319 E7 UniProtKB: P06788 SEQ ID NOG 19320 E7 UniProtKB: P17387 SEQ ID NOG20321 E7 UniProtKB: P06429 SEQ ID NO:321322 E7 UniProtKB: P27230 SEQ ID NO:322323 E7 UniProtKB: P24837 SEQ ID NO:323324 E7 UniProtKB: P21736 SEQ ID NO:324325 E7 UniProtKB: P26558 SEQ ID NO:325326 E7 UniProtKB: P36831 SEQ ID NO:326327 E7 UniProtKB: P36833 SEQ ID NO:327328 E7 UniProtKB: Q9QCZ1 SEQ ID NO:328329 E7 UniProtKB: Q81965 SEQ ID NO:329330 E7 UniProtKB: Q80956 SEQ ID NOG30331 EF1A2 UniProtKB: Q05639 SEQ ID NO:331332 EFTUD2/m UniProtKB: QI5029 SEQ ID NO:332333 EGFR UniProtKB: A0A0B4J1Y5 SEQ ID NO:333334 EGFR UniProtKB: E7BSV0 SEQ ID NO:334335 EGFR UniProtKB: L0R6G1 SEQ ID NO:335336 EGFR UniProtKB: P00533-2 SEQ ID NO:336337 EGFR UniProtKB: P00533 SEQ ID NO:337338 EGFR UniProtKB: Q147T7 SEQ ID NO:338 W O 2018/204534 PCT/US2018/030725 339 EGFR UniProtKB: Q504U8 SEQ ID NO:339340 EGFR UniProtKB: Q8NDU8 SEQ ID NO:340341 EGLN3 UniProtKB: Q9H6Z9 SEQ ID NO: 341342 ELF2/m UniProtKB: B7Z720 SEQ ID NO:342343 EMMPRIN UniProtKB: Q54A51 SEQ ID NO:343344 EpCam UniProtKB: P16422 SEQ ID NO:344345 EphA2 UniProtKB: P29317 SEQ ID NO:345346 EphA3 UniProtKB: P29320 SEQ ID NO:346347 EphA3 UniProtKB: Q6P4R6 SEQ ID NO:347348 ErbB3 UniProtKB: B3KWG5 SEQ ID NO:348349 ErbB3 UniProtKB: B4DGQ7 SEQ ID NO:349350 ERBB4 UniProtKB: Q15303 SEQ ID NO:350351 ERG UniProtKB: Pl 1308 SEQ ID NO:351352 ETV6 UniProtKB: P41212 SEQ ID NO:352353 EWS UniProtKB: Q01844 SEQ ID NO:353354 EZH2 UniProtKB: F2YMM1 SEQ ID NO:354355 EZH2 UniProtKB: G3XAL2 SEQ ID NO:355356 EZH2 UniProtKB: L0R855 SEQ ID NO:356357 EZH2 UniProtKB: Q15910 SEQ ID NO:357358 EZH2 UniProtKB: S4S3R8 SEQ ID NO:358359 FABP7 UniProtKB: 015540 SEQ ID NO:359360 FCGR3 A_V ersion_ 1 UniProtKB: P08637 SEQ ID NO:360361 FCGR3 A_V ersion_2 CCDS: CCDS 1232.1 SEQ ID NO:361362 FGF5 UniProtKB: P12034 SEQ ID NO:362363 FGF5 UniProtKB: Q60518 SEQ ID NO:363364 FGFR2 UniProtKB: P21802 SEQ ID NO:364365 fibronectin UniProtKB: A0A024R5I6 SEQ ID NO:365366 fibronectin UniProtKB: A0A024RB01 SEQ ID NO:366367 fibronectin UniProtKB: A0A024RDT9 SEQ ID NO:367368 fibronectin UniProtKB: A0A024RDV5 SEQ ID NO:368369 fibronectin UniProtKB: A6NH44 SEQ ID NO:369370 fibronectin UniProtKB: A8K6A5 SEQ ID NO:370371 fibronectin UniProtKB: B2R627 SEQ ID NO:371372 fibronectin UniProtKB: B3KXM5 SEQ ID NO:372373 fibronectin UniProtKB: B4DIC5 SEQ ID NO:373374 fibronectin UniProtKB: B4DN21 SEQ ID NO:374375 fibronectin UniProtKB: B4DS98 SEQ ID NO:375376 fibronectin UniProtKB: B4DTH2 SEQ ID NO:376 W O 2018/204534 PCT/US2018/030725 377 fibronectin UniProtKB: B4DTK1 SEQ ID NO:377378 fibronectin UniProtKB: B4DU16 SEQ ID NO:378379 fibronectin UniProtKB: B7Z3W5 SEQ ID NO:379380 fibronectin UniProtKB: B7Z939 SEQ ID NO:380381 fibronectin UniProtKB: G5E9X3 SEQ ID NO:381382 fibronectin UniProtKB: Q9H382 SEQ ID NO:382383 FOS UniProtKB: P01100 SEQ ID NO:383384 FOXP3 UniProtKB: Q9BZS1 SEQ ID NO:384385 FUT1 UniProtKB: P19526 SEQ ID NO:385386 G250 UniProtKB: QI6790 SEQ ID NO:386387 GAGE-1 Genbank: AAA82744 SEQ ID NO:387388 GAGE-2 UniProtKB: Q6NT46 SEQ ID NO:388389 GAGE-3 UniProtKB: QI3067 SEQ ID NO:389390 GAGE-4 UniProtKB: QI3068 SEQ ID NO:390391 GAGE-5 UniProtKB: QI3069 SEQ ID NO:391392 GAGE-6 UniProtKB: QI3070 SEQ ID NO:392393 GAGE7b UniProtKB: 076087 SEQ ID NO:393394 GAGE-8_(GAGE-2D) UniProtKB: Q9UEU5 SEQ ID NO:394395 GASR UniProtKB: P32239 SEQ ID NO:395396 GnT-V UniProtKB: Q09328 SEQ ID NO:396397 GPC3 UniProtKB: I6QJG3 SEQ ID NO:397398 GPC3 UniProtKB: P51654 SEQ ID NO:398399 GPC3 UniProtKB: Q8IYG2 SEQ ID NO:399400 GPNMB/m UniProtKB: A0A024RA55 SEQ ID NO:400401 GPNMB/m UniProtKB: QI4956 SEQ ID NO:401402 GPNMB/m UniProtKB: Q8IXJ5 SEQ ID NO:402403 GPNMB/m UniProtKB: Q96F58 SEQ ID NO:403404 GRM3 UniProtKB: Q14832 SEQ ID NO:404405 HAGE UniProtKB: Q9NXZ2 SEQ ID NO:405406 hepsin UniProtKB: B2ZDQ2 SEQ ID NO:406407 hepsin UniProtKB: P05981 SEQ ID NO:407408 Her2/neu UniProtKB: B4DTR1 SEQ ID NO:408409 Her2/neu UniProtKB: L8E8G2 SEQ ID NO:409410 Her2/neu UniProtKB: P04626 SEQ ID NO:410411 Her2/neu UniProtKB: Q9UK79 SEQ ID NO:411412 HLA-A2/m UniProtKB: Q95387 SEQ ID NO:412413 HLA-A2/m UniProtKB: Q9MYF8 SEQ ID NO:413414 homeobox_NKX3.1 UniProtKB: Q99801 SEQ ID NO:414 W O 2018/204534 PCT/US2018/030725 415 HOM-TES-85 UniProtKB: B2RBQ6 SEQ ID NO:415416 HOM-TES-85 UniProtKB: Q9P127 SEQ ID NO:416417 HPG1 Pubmed: 12543784 SEQ ID NO:417418 HS71A UniProtKB: P0DMV8 SEQ ID NO:418419 HS71B UniProtKB: P0DMV9 SEQ ID NO:419420 HST-2 UniProtKB: P10767 SEQ ID NO:420421 hTERT UniProtKB: 094807 SEQ ID NO:421422 ICE UniProtKB: 000748 SEQ ID NO:422423 IF2B3 UniProtKB: 000425 SEQ ID NO:423424 IL-13Ra2 UniProtKB: QI4627 SEQ ID NO:424425 IL2-RA UniProtKB: P01589 SEQ ID NO:425426 IL2-RB UniProtKB: P14784 SEQ ID NO:426427 IL2-RG UniProtKB: P31785 SEQ ID NO:427428 IMP3 UniProtKB: Q9NV31 SEQ ID NO:428429 ITA5 UniProtKB: P08648 SEQ ID NO:429430 ITB1 UniProtKB: P05556 SEQ ID NO:430431 ITB6 UniProtKB: Pl8564 SEQ ID NO:431432 kallikrein-2 UniProtKB: A0A024R4J4 SEQ ID NO:432433 kallikrein-2 UniProtKB: A0A024R4N3 SEQ ID NO:433434 kallikrein-2 UniProtKB: B0AZU9 SEQ ID NO:434435 kallikrein-2 UniProtKB: B4DU77 SEQ ID NO:435436 kallikrein-2 UniProtKB: P20151 SEQ ID NO:436437 kallikrein-2 UniProtKB: Q6T774 SEQ ID NO:437438 kallikrein-2 UniProtKB: Q6T775 SEQ ID NO:438439 kallikrein-4 UniProtKB: A0A0C4DFQ5 SEQ ID NO:439440 kallikrein-4 UniProtKB: Q5BQA0 SEQ ID NO:440441 kallikrein-4 UniProtKB: Q96PT0 SEQ ID NO:441442 kallikrein-4 UniProtKB: Q96PT1 SEQ ID NO:442443 kallikrein-4 UniProtKB: Q9Y5K2 SEQ ID NO:443444 KI20A UniProtKB: 095235 SEQ ID NO:444445 KIAA0205 UniProtKB: Q92604 SEQ ID NO:445446 KIF2C UniProtKB: Q99661 SEQ ID NO:446447 KK-LC-] UniProtKB: Q5H943 SEQ ID NO:447448 EDER UniProtKB: P01130 SEQ ID NO:448449 LGMN UniProtKB: Q99538 SEQ ID NO:449450 LIRB2 UniProtKB: Q8N423 SEQ ID NO:450451 LY6K UniProtKB: Q17RY6 SEQ ID NO:451452 MAGAS UniProtKB: P43359 SEQ ID NO:452 W O 2018/204534 PCT/US2018/030725 453 MAGAS UniProtKB: P43361 SEQ ID NO:453454 MAGAB UniProtKB: P43364 SEQ ID NO:454455 MAGE-A10 UniProtKB: A0A024RC14 SEQ ID NO:455456 MAGE-A12 UniProtKB: P43365 SEQ ID NO:456457 MAGE-A1 UniProtKB: P43355 SEQ ID NO:457458 MAGE-A2 UniProtKB: P43356 SEQ ID NO:458459 MAGE-A3 UniProtKB: P43357 SEQ ID NO:459460 MAGE-A4 UniProtKB: A0A024RC12 SEQ ID NO:460461 MAGE-A4 UniProtKB: P43358 SEQ ID NO:461462 MAGE-A4 UniProtKB: Q1RN33 SEQ ID NO:462463 MAGE-A6 UniProtKB: A8K072 SEQ ID NO:463464 MAGE-A6 UniProtKB: P43360 SEQ ID NO:464465 MAGE-A6 UniProtKB: Q6FHI5 SEQ ID NO:465466 MAGE-A9 UniProtKB: P43362 SEQ ID NO:466467 MAGE-BIO UniProtKB: Q96LZ2 SEQ ID NO:467468 MAGE-B16 UniProtKB: A2A368 SEQ ID NO:468469 MAGE-B17 UniProtKB: A8MXT2 SEQ ID NO:469470 MAGE-_B1 UniProtKB: Q96TG1 SEQ ID NO:470471 MAGE-B2 UniProtKB: 015479 SEQ ID NO:471472 MAGE-B3 UniProtKB: 015480 SEQ ID NO:472473 MAGE-B4 UniProtKB: 015481 SEQ ID NO:473474 MAGE-B5 UniProtKB: Q9BZ81 SEQ ID NO:474475 MAGE-B6 UniProtKB: Q8N7X4 SEQ ID NO:475476 MAGE-CI UniProtKB: 060732 SEQ ID NO:476477 MAGE-C2 UniProtKB: Q9UBF1 SEQ ID NO:477478 MAGE-C3 UniProtKB: Q8TD91 SEQ ID NO:478479 MAGE-D1 UniProtKB: Q9Y5V3 SEQ ID NO:479480 MAGE-D2 UniProtKB: Q9UNF1 SEQ ID NO:480481 MAGE-D4 UniProtKB: Q96JG8 SEQ ID NO:481482 MAGE-_E1 UniProtKB: Q6IAI7 SEQ ID NO:482483 MAGE-E1_(MAGE1) UniProtKB: Q9HCI5 SEQ ID NO:483484 MAGE-E2 UniProtKB: Q8TD90 SEQ ID NO:484485 MAGE-F1 UniProtKB: Q9HAY2 SEQ ID NO:485486 MAGE-H1 UniProtKB: Q9H213 SEQ ID NO:486487 MAGEL2 UniProtKB: Q9UJ55 SEQ ID NO:487488 mammaglobin_A UniProtKB: QI3296 SEQ ID NO:488489 mammaglobin_A UniProtKB: Q6NX70 SEQ ID NO:489490 MART-l/melan-A UniProtKB: QI6655 SEQ ID NO:490 W O 2018/204534 PCT/US2018/030725 WO 2018/204534 PCT/US2018/030725 rq m in oo CT O rq n ،־ oo CT O rq cn >כ oo CT o rq cn in oo cr cr cr cr. cr cr T ؛ cr CT O o o o o o O O o o H H H H H H ז— H H H H rq rq rq rq rq rq rq rq rq י؛ י؛ י؛ י؛ י؛ י؛ י؛ V) V) V) V) V) V) V) V) V) V) m m m m m m m m in in V) V) V) V) V) V) V) V) V) d d d d d d d d d d d d d o o o o o o o o o o o o o o o o o o o o o o o o oz z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z z Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q Q QO' O' O' O' O' O' O' O' O' O' O' O' o O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O' O'm m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO OO Ox o rq cn m rq m oo CT o rq in oo o rq cn rq oo >כ>- rq ־ן ।§ § § § § CT CT CT CT CT CT CT כ o H D I—»b I—»b I—» JU 1D D D D DDDDDDDDD >כ m O 1Q 61Q 7O' Q8:1Q 9'Q9Q9 I 1 I 1 I 1 I 1>כ >כ >כ >כ >כ >כ >כ >כ CT CT CT CT CT CT CT CT CT CT CT CT CT CT CT CT CT oOo o o oO' O O O O o' o o o o o O' o o؛<מmm m m mm m m m m m m m m m m m מ mom m m m mm m m מ מ m m מ m m מ m o ob b b 6b b b b b b b b b b b b o o c c c c o c c b o o c b b o b b o b£ % ־S £ ־S£ ־S£ ־S niP£ 'a£ 'a£ 'a£ 'a£ 'a n iP in iP in iP in iP in iP in iP in iP i£ £niPniPniPniP£־niPniPn iP i£ ££n iP i£ c£n iP in iP i £ £ 'aDDD D D הD D D D D D D D D D D DD Dק ה ה ק D ה ק D D DD DDD DD D rq H 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 01 Cd u u u u ClClClClu ClClClClClClClClClu ClClClClClClClClClu u Clu ClClClClu2 2 2 2 2 2 2 2 2 2 S S S S S rq m m OO CT o rq n ،־ OO CT o rq cn >כ OO CT o rq cn in OO cr cr. cr. cr. cr. CT. cr. cr. CT o o o o o o O o o O H H H H H H ז— H H H H rq rq rq rq rq rq rq rq rq V) V) V) V) V) V) V) V) V) V) in in in V) V) V) V) V) V) V) V) V) 529 MC1_R UniProtKB: V9QDN7 SEQ ID NO:529530 MC1_R UniProtKB: V9QDQ6 SEQ ID NO:530531 mesothelin UniProtKB: Q13421 SEQ ID NO:531532 MITF UniProtKB: 075030-8 SEQ ID NO:532533 MITF UniProtKB: 075030-9 SEQ ID NO:533534 MITF UniProtKB: 075030 SEQ ID NO:534535 MMP1_1 UniProtKB: B3KQS8 SEQ ID NO:535536 MMP7 UniProtKB: P09237 SEQ ID NO:536537 MUC-1 Genbank: AAA60019 SEQ ID NO:537538 MUM-l/m RefSeq: NP_116242 SEQ ID NO:538539 MUM-2/m UniProtKB: Q9Y5R8 SEQ ID NO:539540 MYO1A UniProtKB: Q9UBC5 SEQ ID NO:540541 MYO IB UniProtKB: 043795 SEQ ID NO:541542 MYO IC UniProtKB: 000159 SEQ ID NO:542543 MYOID UniProtKB: 094832 SEQ ID NO:543544 MYO IE UniProtKB: QI2965 SEQ ID NO:544545 MYO IF UniProtKB: 000160 SEQ ID NO:545546 MYO1G UniProtKB: B0I1T2 SEQ ID NO:546547 MYO1H RefSeq: NP_001094891 SEQ ID NO:547548 NA17 UniProtKB: Q3V5L5 SEQ ID NO:548549 NA88-A Pubmed: 10790436 SEQ ID NO:549550 Neo-PAP UniProtKB: Q9BWT3 SEQ ID NO:550551 NFYC/m UniProtKB: QI3952 SEQ ID NO:551552 NGEP UniProtKB: Q6IWH7 SEQ ID NO:552553 NPM UniProtKB: P06748 SEQ ID NO:553554 NRCAM UniProtKB: Q92823 SEQ ID NO:554555 NSE UniProtKB: P09104 SEQ ID NO:555556 NUF2 UniProtKB: Q9BZD4 SEQ ID NO:556557 NY-ESO-1 UniProtKB: P78358 SEQ ID NO:557558 OA1 UniProtKB: P51810 SEQ ID NO:558559 OGT UniProtKB: 015294 SEQ ID NO:559560 OS-9 UniProtKB: B4DH11 SEQ ID NO:560561 OS-9 UniProtKB: B4E321 SEQ ID NO:561562 OS-9 UniProtKB: B7Z8E7 SEQ ID NO:562563 OS-9 UniProtKB: QI3438 SEQ ID NO:563564 osteocalcin UniProtKB: P02818 SEQ ID NO:564565 osteopontin UniProtKB: A0A024RDE2 SEQ ID NO:565566 osteopontin UniProtKB: A0A024RDE6 SEQ ID NO:566 W O 2018/204534 PCT/US2018/030725 567 osteopontin UniProtKB: A0A024RDJ0 SEQ ID NO:567568 osteopontin UniProtKB: B7Z351 SEQ ID NO:568569 osteopontin UniProtKB: F2YQ21 SEQ ID NO:569570 osteopontin UniProtKB: P10451 SEQ ID NO:570571 p53 UniProtKB: P04637 SEQ ID NO:571572 PAGE-4 UniProtKB: 060829 SEQ ID NO:572573 PAI-1 UniProtKB: P05121 SEQ ID NO:573574 PAI-2 UniProtKB: P05120 SEQ ID NO:574575 PAP UniProtKB: Q06141 SEQ ID NO:575576 PAP UniProtKB: Q53S56 SEQ ID NO:576577 PATE UniProtKB: Q8WXA2 SEQ ID NO:577578 PAX3 UniProtKB: P23760 SEQ ID NO:578579 PAX5 UniProtKB: Q02548 SEQ ID NO:579580 PD1L1 UniProtKB: Q9NZQ7 SEQ ID NO:580581 PDCD1 UniProtKB: Q15116 SEQ ID NO:581582 PDEF UniProtKB: 095238 SEQ ID NO:582583 PEC Al UniProtKB: P16284 SEQ ID NO:583584 PGCB UniProtKB: Q96GW7 SEQ ID NO:584585 PGFRB UniProtKB: P09619 SEQ ID NO:585586 Pim-1_-Kinase UniProtKB: A0A024RD25 SEQ ID NO:586587 Pin-1 UniProtKB: 015428 SEQ ID NO:587588 Pin-1 UniProtKB: QI3526 SEQ ID NO:588589 Pin-1 UniProtKB: Q49AR7 SEQ ID NO:589590 PLAC1 UniProtKB: Q9HBJ0 SEQ ID NO:590591 PMEL UniProtKB: P40967 SEQ ID NO:591592 PML UniProtKB: P29590 SEQ ID NO:592593 POTEF UniProtKB: A5A3E0 SEQ ID NO:593594 POTE UniProtKB: Q86YR6 SEQ ID NO:594595 FRAME UniProtKB: A0A024R1E6 SEQ ID NO:595596 FRAME UniProtKB: P78395 SEQ ID NO:596597 PRDX5/m UniProtKB: P30044 SEQ ID NO:597598 PRM2 UniProtKB: P04554 SEQ ID NO:598599 prostein UniProtKB: Q96JT2 SEQ ID NO:599600 proteinase-3 UniProtKB: D6CHE9 SEQ ID NO:600601 proteinase-3 UniProtKB: P24158 SEQ ID NO:601602 PSA UniProtKB: P55786 SEQ ID NO:602603 PSB9 UniProtKB: P28065 SEQ ID NO:603604 PSCA UniProtKB: D3DWI6 SEQ ID NO:604 W O 2018/204534 PCT/US2018/030725 605 PSCA UniProtKB: 043653 SEQ ID NO:605606 PSGR UniProtKB: Q9H255 SEQ ID NO:606607 PSM UniProtKB: Q04609 SEQ ID NO:607608 PTPRC RefSeq: NP_002829 SEQ ID NO:608609 RAB8A UniProtKB: P61006 SEQ ID NO:609610 RAGE-1 UniProtKB: Q9UQ07 SEQ ID NO:610611 RARA UniProtKB: P10276 SEQ ID NO:611612 RASH UniProtKB: P01112 SEQ ID NO:612613 RASK UniProtKB: P01116 SEQ ID NO:613614 RASN UniProtKB: P01111 SEQ ID NO:614615 RGS5 UniProtKB: 015539 SEQ ID NO:615616 RHAMM/CD168 UniProtKB: 075330 SEQ ID NO:616617 RHOC UniProtKB: P08134 SEQ ID NO:617618 RSSA UniProtKB: P08865 SEQ ID NO:618619 RUI UniProtKB: Q9UHJ3 SEQ ID NO:619620 RU2 UniProtKB: Q9UHG0 SEQ ID NO:620621 RUNX1 UniProtKB: Q01196 SEQ ID NO:621622 S-100 UniProtKB: V9HW39 SEQ ID NO:622623 SAGE UniProtKB: Q9NXZ1 SEQ ID NO:623624 SART-_1 UniProtKB: 043290 SEQ ID NO:624625 S ART-2 UniProtKB: Q9UL01 SEQ ID NO:625626 S ART-3 UniProtKB: QI5020 SEQ ID NO:626627 SEPR UniProtKB: Q12884 SEQ ID NO:627628 SIA7F UniProtKB: Q969X2 SEQ ID NO:628629 SIA8A UniProtKB: Q92185 SEQ ID NO:629630 SIAT9 UniProtKB: Q9UNP4 SEQ ID NO:630631 SIRT2/m UniProtKB: A0A024R0G8 SEQ ID NO:631632 SIRT2/m UniProtKB: Q8IXJ6 SEQ ID NO:632633 SOX 10 UniProtKB: P56693 SEQ ID NO:633634 SP17 UniProtKB: QI5506 SEQ ID NO:634635 SPNXA UniProtKB: Q9NS26 SEQ ID NO:635636 SPXN3 UniProtKB: Q5MJ09 SEQ ID NO:636637 SSX-1 UniProtKB: Q16384 SEQ ID NO:637638 SSX-2 UniProtKB: Q16385 SEQ ID NO:638639 SSX3 UniProtKB: Q99909 SEQ ID NO:639640 SSX-4 UniProtKB: 060224 SEQ ID NO:640641 ST1A1 UniProtKB: P50225 SEQ ID NO:641642 STAG2 UniProtKB: Q8N3U4-2 SEQ ID NO:642 W O 2018/204534 PCT/US2018/030725 643 STAMP-1 UniProtKB: Q8NFT2 SEQ ID NO:643644 STEAP-1 UniProtKB: A0A024RA63 SEQ ID NO:644645 STEAP-1 UniProtKB: Q9UHE8 SEQ ID NO:645646 Survivin-2B UniProtKB: 015392-2 SEQ ID NO:646647 survivin UniProtKB: 015392 SEQ ID NO:647648 SYCPl UniProtKB: A0A024R0I2 SEQ ID NO:648649 SYCP1 UniProtKB: B7ZLS9 SEQ ID NO:649650 SYCPl UniProtKB: QI5431 SEQ ID NO:650651 SYCPl UniProtKB: Q3MHC4 SEQ ID NO:651652 SYT-SSX-1 UniProtKB: A4PIV7 SEQ ID NO:652653 SYT-SSX-1 UniProtKB: A4PIV8 SEQ ID NO:653654 SYT-SSX-2 UniProtKB: A4PIV9 SEQ ID NO:654655 SYT-SSX-2 UniProtKB: A4PIW0 SEQ ID NO:655656 TARP UniProtKB: Q0VGM3 SEQ ID NO:656657 TCRg UniProtKB: A2JGV3 SEQ ID NO:657658 TF2AA UniProtKB: P52655 SEQ ID NO:658659 TGFR2 UniProtKB: P37173 SEQ ID NO:659660 TGM-4 UniProtKB: B2R7D1 SEQ ID NO:660661 TIE2 UniProtKB: Q02763 SEQ ID NO:661662 TKTL1 UniProtKB: P51854 SEQ ID NO:662663 TPI/m UniProtKB: P60174 SEQ ID NO:663664 TRGV11 UniProtKB: Q99601 SEQ ID NO:664665 TRGV9 UniProtKB: A4D1X2 SEQ ID NO:665666 TRGV9 UniProtKB: Q99603 SEQ ID NO:666667 TRGV9 UniProtKB: Q99604 SEQ ID NO:667668 TRPC1 UniProtKB: P48995 SEQ ID NO:668669 TRP-p8 UniProtKB: Q7Z2W7 SEQ ID NO:669670 TSG10 UniProtKB: Q9BZW7 SEQ ID NO:670671 TSPY1 UniProtKB: Q01534 SEQ ID NO:671672 TVC_(TRGV3) Genbank: Ml3231.1 SEQ ID NO:672673 TX101 UniProtKB: Q9BY14-2 SEQ ID NO:673674 tyrosinase UniProtKB: A0A024DBG7 SEQ ID NO:674675 tyrosinase UniProtKB: L8B082 SEQ ID NO:675676 tyrosinase UniProtKB: L8B086 SEQ ID NO:676677 tyrosinase UniProtKB: L8B0B9 SEQ ID NO:677678 tyrosinase UniProtKB: 075767 SEQ ID NO:678679 tyrosinase UniProtKB: P14679 SEQ ID NO:679680 tyrosinase UniProtKB: U3M8N0 SEQ ID NO:680 W O 2018/204534 PCT/US2018/030725 681 tyrosinase UniProtKB: U3M9D5 SEQID NO:681682 tyrosinase UniProtKB: U3M9J2 SEQ ID NO:682683 TYRP1 UniProtKB: P17643 SEQ ID NO:683684 TYRP2 UniProtKB: P40126 SEQ ID NO:684685 UFA UniProtKB: Q96NZ9 SEQ ID NO:685686 VEGFR1 UniProtKB: B5A924 SEQ ID NO:686687 WT1 UniProtKB: A0A0H5AUY0 SEQ ID NO:687688 WT1 UniProtKB: P19544 SEQ ID NO:688689 WT1 UniProtKB: Q06250 SEQ ID NO:689690 XAGE1 UniProtKB: Q9HD64 SEQ ID NO:690691 IL-10 UniProtKB: P22301 SEQID NO:691692 IL-5 UniProtKB: P05113 SEQ ID NO:692693 M-CSF UniProtKB: P09603 SEQ ID NO:693694 TGFbetal UniProtKB: P01137 SEQ ID NO:694695 Caspase_8 UniProtKB: QI4790 SEQ ID NO:695696 SERPINB5 UniProtKB: P36952 SEQ ID NO:696697 calreticulin UniProtKB: B4DHR1 SEQ ID NO:697698 calreticulin UniProtKB: B4E2Y9 SEQ ID NO:698699 calreticulin UniProtKB: P27797 SEQ ID NO:699700 calreticulin UniProtKB: Q96L12 SEQ ID NO:700701 N-myc UniProtKB: P04198 SEQ ID NO:701 W O 2018/204534 PCT/US2018/030725 WO 2018/204534 PCT/US2018/030725 id="p-150" id="p-150" id="p-150"
[0150] Additionally, the antigens (including the sequences/fragments/epitopes shown in Column 4) described in Table 1 can be cloned into the LAMP Constructs described herein either individually, or in combination with one another. For example, the names pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT- 1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3 refer to the proteins described in Table 1 and are explicitly intended to refer generically not only to the full-length sequences shown in Column 1 of Table 1, but also, and preferably, the sequences/fragments/epitopes as described in Figures 19 and 20 and in the fourth Column of Table 1. Thus, each one of the sequences shown in Column 1 of Table 1, including the epitopes/fragments described in Column 4 of Table 1 can be used to generate a LAMP Construct. To illustrate different possible combinations, but in no way limiting the disclosure, the combinations of antigens (including the sequences shown in Column 1 of Table 1 and/or the sequences/fragments/epitopes described in Column 4 of Table 1) can be cloned into the LAMP Constructs as follows: (a) pp65 and at least one of gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (b) gB and at least one of pp65, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY- ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (c) IEand at least one of pp65, gB, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (d) MTRII, and at least one of pp65, gB, IE1, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (e) US28 and at least one of pp65, gB, IE1, MTRII, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40- L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (f) IGFBP2 and at least one of pp65, gB, IE1, WO 2018/204534 PCT/US2018/030725 MTRII, US28, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (g) IL 10 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY- ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (h) UL144 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (i) UL141 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (j) US11 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40- L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (k) IE2 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (1) TERT and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, Survivin, Tetanus, NY- ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (m) Survivin and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (n) Tetanus and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, WO 2018/204534 PCT/US2018/030725 HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (o) NY-ESO-1 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40- L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (p) HER2 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (q) HER3 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (r) HVEM and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL 10, ULI44, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (s) HOS and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (t) HPV16E6 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40- L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (u) HPV18E6 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (v) HPV16E7 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, WO 2018/204534 PCT/US2018/030725 BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (w) HPV18E7 and at least one ofpp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (x) EBNA1 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (y) EBNA1 trunc and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40- L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (z) gp350 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (aa) LMP2 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ab) GCP3 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ac) Middle S and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT- 1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ad) X Protein and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ae) TIGIT and at least one of pp65, gB, WO 2018/204534 PCT/US2018/030725 IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (af) TEM8 and at least one ofpp65, gB, IE1, MTRII, US28, IGFBP2, IL 10, UE 144, UE 141, US 11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ag) TEM1 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ah) HER2 ECD+TM and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ai) CEA and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN- iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (aj) TARP and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO- 1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ak) PROSTEIN and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL 10, ULI44, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (al) PSMA and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL 10, ULI44, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT- trunc., FRAME, LAGE-1, and/or MAGE A3; (am) BIRC4 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, WO 2018/204534 PCT/US2018/030725 HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (an) MUCIN-1 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO- 1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ao) MUCIN-iso and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, CD40-L, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ap) CD40-L and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL 10, ULI44, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, WT-1, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (aq) WT-1 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40- L, WT-1 trunc., FRAME, LAGE-1, and/or MAGE A3; (ar) WT-1 trunc and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, FRAME, LAGE-1, and/or MAGE A3; (as) FRAME and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., LAGE-1, and/or MAGE A3; (at) LAGE-1 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL 10, ULI44, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X Protein, TIGIT, TEM8, TEM1, HERECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, and/or MAGE A3; and/or (au) MAGE A3 and at least one of pp65, gB, IE1, MTRII, US28, IGFBP2, IL10, UL144, UL141, US11, HOS, IE2, TERT, Survivin, Tetanus, NY-ESO-1, HER2, HER3, HVEM, HPV16E6, HPV18E6, HPV16E7, HPV18E7, EBNA1, EBNA trunc, gp350, LMP2, GCP3, Middle S, X WO 2018/204534 PCT/US2018/030725 Protein, TIGIT, TEM8, TEM1, HER2 ECD+TM, CEA, TARP, PROSTEIN, PSMA, BIRC4, MUCIN-1, MUCIN-1 iso, CD40-L, WT-1, WT-1 trunc., FRAME, and/or LAGE-1. The order of the combination of antigens as described above in a particular LAMP construct can vary as this list describes what a LAMP Construct comprises and not necessarily to describe the arrangement of the antigens within a particular construct. Moreover, it is specifically envisioned that these antigens can be combined within a single LAMP Construct, or can be delivered in a composition comprising multiple LAMP Constructs.
Assembly of Sequences Encoding Improved LAMP Constructs [0151] Procedures for constructing improved LAMP Constructs comprising the antigen of interest are well known in the art (see e.g., Williams, et al., J. Cell Biol. Ill: 955, 1990). DNA sequences encoding the desired segments can be obtained from readily available recombinant DNA materials such as those available from the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Md. 20852, U.S.A., or from DNA libraries that contain the desired DNA.[0152] For example, the DNA segments corresponding to the desired domain sequences can be assembled with appropriate control and signal sequences using routine procedures of recombinant DNA methodology. See, e.g., as described in U.S. Pat. No. 4,593,002, and Langford, et al., Molec. Cell. Biol. 6: 3191, 1986.[0153] A DNA sequence encoding a protein or polypeptide can be synthesized chemically or isolated by one of several approaches. The DNA sequence to be synthesized can be designed with the appropriate codons for the desired amino acid sequence. In general, one will select preferred codons for the intended host in which the sequence will be used for expression. The complete sequence may be assembled from overlapping oligonucleotides prepared by standard methods and assembled into a complete coding sequence. See, e.g., Edge, Nature 292: 756, 1981; Nambair, et al. Science 223: 1299, 1984; Jay, et al., J. Biol. Chern. 259: 6311, 1984.[0154] In one aspect, one or more of the nucleic acids encoding the domain sequences of the improved LAMP Construct are isolated individually using the polymerase chain reaction (M. A. Innis, et al., In PCR Protocols: A Guide to Methods and Applications, Academic Press, 1990). The domains are preferably isolated from publicly available clones known to contain them, but they may also be isolated from genomic DNA or cDNA libraries. Preferably, isolated fragments are bordered by compatible restriction endonuclease sites which allow an improved LAMP Construct encoding the cancer antigen sequence to be constructed. This technique is well known to those of skill in the art. Domain sequences may be fused directly to each other (e.g., with no intervening sequences), or inserted into one another (e.g., where domain sequences are discontinuous), or may be separated by intervening sequences (e.g., such as linker sequences).[0155] The basic strategies for preparing oligonucleotide primers, probes and DNA libraries, as well as their screening by nucleic acid hybridization, are well known to those of ordinary skill in the art. See, e.g., Sambrook, et al., 1989, supra; Perbal, 1984, supra. The construction of an appropriate genomic DNA or cDNA 50 WO 2018/204534 PCT/US2018/030725 library is within the skill of the art. See, e.g., Perbal, 1984, supra. Alternatively, suitable DNA libraries or publicly available clones are available from suppliers of biological research materials, such as Clonetech and Stratagene, as well as from public depositories such as the American Type Culture Collection.[0156] Selection may be accomplished by expressing sequences from an expression library of DNA and detecting the expressed peptides immunologically. Clones which express peptides that bind to MHC II molecules and to the desired antibodies/T cell receptors are selected. These selection procedures are well known to those of ordinary skill in the art (see, e.g., Sambrook, et al., 1989, supra).[0157] Once a clone containing the coding sequence for the desired polypeptide sequence has been prepared or isolated, the sequence can be cloned into any suitable vector, preferably comprising an origin of replication for maintaining the sequence in a host cell.
Nucleic Acid Delivery Vehicles [0158] In one aspect, a vaccine composition comprising an improved LAMP Construct is introduced into a cell. The cell may be a host cell for replicating the nucleic acid or for expressing the improved LAMP Construct. Preferably, the host cell for expressing the improved LAMP Construct is an antigen presenting cell (described further below).[0159] In preferred embodiments, the improved LAMP Construct further comprises a polynucleotide sequence for insertion into a target cell and an expression control sequence operably linked thereto to control expression of the polynucleotide sequence (e.g., transcription and/or translation) in the cell. Examples include plasmids, phages, autonomously replicating sequences (ARS), centromeres, and other sequences which are able to replicate or be replicated in vitro or in a host cell (e.g., such as a bacterial, yeast, or insect cell) and/or target cell (e.g., such as a mammalian cell, preferably an antigen presenting cell) and/or to convey the sequences encoding the improved LAMP Construct to a desired location within the target cell.[0160] Recombinant expression vectors may be derived from micro-organisms which readily infect animals, including man, horses, cows, pigs, llamas, giraffes, dogs, cats or chickens. Preferred vectors include those which have already been used as live vaccines, such as vaccinia. These recombinants can be directly inoculated into a host, conferring immunity not only to the microbial vector, but also to express foreign antigens. Preferred vectors contemplated herein as live recombinant vaccines include RNA viruses, adenovirus, herpesviruses, poliovirus, and vaccinia and other pox viruses, as taught in Flexner, Adv. Pharmacol. 21: 51, 1990, for example.[0161] Expression control sequences include, but are not limited to, promoter sequences to bind RNA polymerase, enhancer sequences or negative regulatory elements to bind to transcriptional activators and repressors, respectively, and/or translation initiation sequences for ribosome binding. For example, a bacterial expression vector can include a promoter such as the lac promoter and for transcription initiation, the Shine- Dalgarno sequence and the start codon AUG (Sambrook, et al., 1989, supra). Similarly, a eukaryotic expression WO 2018/204534 PCT/US2018/030725 vector preferably includes a heterologous, homologous, or chimeric promoter for RNA polymerase II, a downstream polyadenylation signal, the start codon AUG, and a termination codon for detachment of a ribosome.[0162] Expression control sequences may be obtained from naturally occurring genes or may be designed. Designed expression control sequences include, but are not limited to, mutated and/or chimeric expression control sequences or synthetic or cloned consensus sequences. Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are well known in the art. Such vectors are capable of transcribing RNA in vitro or in vivo, and are commercially available from sources such as Stratagene (La Jolla, Calif.) and Promega Biotech (Madison, Wis.).[0163] In order to optimize expression and/or transcription, it may be necessary to remove, add or alter 5' and/or 3' untranslated portions of the vectors to eliminate extra, or alternative translation initiation codons or other sequences that may interfere with, or reduce, expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites can be inserted immediately 5' of the start codon to enhance expression. A wide variety of expression control sequences—sequences that control the expression of a DNA sequence operatively linked to it—may be used in these vectors to express the DNA sequences of this invention. Such useful expression control sequences include, for example, the early or late promoters of SV40, CMV, vaccinia, polyoma, adenovirus, herpes virus and other sequences known to control the expression of genes of mammalian cells, and various combinations thereof.[0164] In one aspect, the improved LAMP Construct comprises an origin of replication for replicating the vector. Preferably, the origin functions in at least one type of host cell which can be used to generate sufficient numbers of copies of the sequence for use in delivery to a target cell. Suitable origins therefore include, but are not limited to, those which function in bacterial cells (e.g., such as Escherichia sp., Salmonella sp., Proteus sp., Clostridium sp., Klebsiella sp., Bacillus sp., Streptomyces sp., and Pseudomonas sp.), yeast (e.g., such as Saccharamyces sp. or Pichia sp.), insect cells, and mammalian cells. In one preferred aspect, an origin of replication is provided which functions in the target cell into which the nucleic acid delivery vehicle is introduced (e.g., a mammalian cell, such as a human cell). In another aspect, at least two origins of replication are provided, one that functions in a host cell and one that functions in a target cell.[0165] The improved LAMP Construct may alternatively, or additionally, comprise sequences to facilitate integration of at least a portion of the nucleic acid deliver vector into a target cell chromosome. For example, the improved LAMP Construct may comprise regions of homology to target cell chromosomal DNA. In one aspect, the delivery vector comprises two or more recombination sites which flank a nucleic acid sequence encoding the improved LAMP Construct.[0166] The vector may additionally comprise a detectable and/or selectable marker to verify that the vector has been successfully introduced in a target cell and/or can be expressed by the target cell. These markers WO 2018/204534 PCT/US2018/030725 can encode an activity, such as, but not limited to, production of RNA, peptide, or protein, or can provide a binding site for RNA, peptides, proteins, inorganic and organic compounds or compositions and the like.[0167] Examples of detectable/selectable markers genes include, but are not limited to: DNA segments that encode products which provide resistance against otherwise toxic compounds (e.g., antibiotics); DNA segments that encode products which are otherwise lacking in the recipient cell (e.g., tRNA genes, auxotrophic markers); DNA segments that encode products which suppress the activity of a gene product; DNA segments that encode products which can be readily identified (e.g., phenotypic markers such as beta-galactosidase, a fluorescent protein (GFP, CEP, YFG, BFP, RFP, EGFP, EYFP, EBFP, dsRed, mutated, modified, or enhanced forms thereof, and the like), and cell surface proteins); DNA segments that bind products which are otherwise detrimental to cell survival and/or function; DNA segments that otherwise inhibit the activity of other nucleic acid segments (e.g., antisense oligonucleotides); DNA segments that bind products that modify a substrate (e.g., restriction endonucleases); DNA segments that can be used to isolate or identify a desired molecule (e.g., segments encoding specific protein binding sites); primer sequences; DNA segments, which when absent, directly or indirectly confer resistance or sensitivity to particular compounds; and/or DNA segments that encode products which are toxic in recipient cells.[0168] The marker gene can be used as a marker for conformation of successful gene transfer and/or to isolate cells expressing transferred genes and/or to recover transferred genes from a cell. For example, in one aspect, the marker gene is used to isolate and purify antigen presenting cells expressing the improved LAMP Constructs.[0169] Substantially similar genes may be provided, e.g., genes with greater than about 50%, greater than about 70%, greater than 80%, greater than about 90%, and preferably, greater than about 95% identity to a known gene. Substantially similar domain sequences may initially be identified by selecting a sequence which specifically hybridizes to a domain sequence of interest under stringent hybridization conditions. Performing assays to determine the suitability of homologous, variant, or modified domain sequences is merely a matter of screening for sequences which express the appropriate activity. Such screening is routine in the art.[0170] The improved LAMP Construct may be provided as naked nucleic acids or in a delivery vehicle associated with one or more molecules for facilitating entry of a nucleic acid into a cell. Suitable delivery vehicles include, but are not limited to: liposomal formulations, polypeptides, polysaccharides, lipopolysaccharides, viral formulations (e.g., including viruses, viral particles, artificial viral envelopes and the like), cell delivery vehicles, and the like.
Lipid-Based Formulations [0171] Delivery vehicles designed to facilitate intracellular delivery of the improved LAMP Constructs must interact with both non-polar and polar environments (in or on, for example, the plasma membrane, tissue fluids, compartments within the cell, and the like). Therefore, preferably, delivery vehicles WO 2018/204534 PCT/US2018/030725 are designed to contain both polar and non-polar domains or a translocating sequence for translocating an improved LAMP Construct into a cell.[0172] Compounds having polar and non-polar domains are termed amphiphiles. Cationic amphiphiles have polar groups that are capable of being positively charged at, or around, physiological pH for interacting with negatively charged polynucleotides such as DNA.[0173] The improved LAMP Constructs described herein can be provided in formulations comprising lipid monolayers or bilayers to facilitate transfer of the vectors across a cell membrane. Liposomes or any form of lipid membrane, such as planar lipid membranes or the cell membrane of an intact cell, e.g., a red blood cell, can be used. Liposomal formulations can be administered by any means, including administration intravenously or orally.[0174] Liposomes and liposomal formulations can be prepared according to standard methods and are well known in the art, see, e.g., Remington's; Akimaru, 1995, Cytokines Mol. Ther. 1: 197-210; Alving, 1995, Immunol. Rev. 145: 5-31; Szoka, 1980, Ann. Rev. Biophys. Bioeng. 9: 467; U.S. Pat. No. 4,235,871; U.S. Pat. No. 4,501,728; and U.S. Pat. No. 4,837,028. In one aspect, the liposome comprises a targeting molecule for targeting a liposome :improved LAMP Construct complex to a particular cell type. In a particularly preferred aspect, a targeting molecule comprises a binding partner (e.g., a ligand or receptor) for a biomolecule (e.g., a receptor or ligand) on the surface of a blood vessel or a cell found in a target tissue.[0175] Liposome charge is an important determinant in liposome clearance from the blood, with negatively charged liposomes being taken up more rapidly by the reticuloendothelial system (Juliano, 1975, Biochem. Biophys. Res. Commun. 63: 651) and thus having shorter half-lives in the bloodstream. Incorporating phosphatidylethanolamine derivatives enhances the circulation time by preventing liposomal aggregation. For example, incorporation of N-(omega-carboxy)acylamidophosphatidylethanolamines into large unilamellar vesicles of L-alpha-distearoylphosphatidylcholine dramatically increases the in vivo liposomal circulation lifetime (see, e.g., Ahl, 1997, Biochim. Biophys. Acta 1329: 370-382). Liposomes with prolonged circulation half-lives are typically desirable for therapeutic and diagnostic uses. For a general discussion of pharmacokinetics, see, e.g., Remington's, Chapters 37-39, Lee, et al., In Pharmacokinetic Analysis: A Practical Approach (Technomic Publishing AG, Basel, Switzerland 1996).[0176] Typically, liposomes are prepared with about 5 to 15 mole percent negatively charged phospholipids, such as phosphatidylglycerol, phosphatidylserine or phosphatidyl-inositol. Added negatively charged phospholipids, such as phosphatidylglycerol, also serve to prevent spontaneous liposome aggregation, and thus minimize the risk of undersized liposomal aggregate formation. Membrane-rigidifying agents, such as sphingomyelin or a saturated neutral phospholipid, at a concentration of at least about 50 mole percent, and to 15 mole percent of monosialylganglioside can also impart desirably liposome properties, such as rigidity (see, e.g., U.S. Pat. No. 4,837,028).
WO 2018/204534 PCT/US2018/030725 id="p-177" id="p-177" id="p-177"
[0177] Additionally, the liposome suspension can include lipid-protective agents which protect lipids against free-radical and lipid-peroxidative damages on storage. Lipophilic free-radical quenchers, such as alpha- tocopherol and water-soluble iron-specific chelators, such as ferrioxianine, are preferred.[0178] The improved LAMP Constructs of the invention can include multilamellar vesicles of heterogeneous sizes. For example, vesicle-forming lipids can be dissolved in a suitable organic solvent or solvent system and dried under vacuum or an inert gas to form a thin lipid film. If desired, the film can be redissolved in a suitable solvent, such as tertiary butanol, and then lyophilized to form a more homogeneous lipid mixture which is in a more easily hydrated powderlike form. This film is covered with an aqueous solution of the peptide or polypeptide complex and allowed to hydrate, typically over a 15 to 60 minute period with agitation. The size distribution of the resulting multilamellar vesicles can be shifted toward smaller sizes by hydrating the lipids under more vigorous agitation conditions or by adding solubilizing detergents such as deoxycholate. The hydration medium preferably comprises the nucleic acid at a concentration which is desired in the interior volume of the liposomes in the final liposome suspension.[0179] Following liposome preparation, the liposomes can be sized to achieve a desired size range and relatively narrow distribution of liposome sizes. One preferred size range is about 0.2 to 0.4 microns, which allows the liposome suspension to be sterilized by filtration through a conventional filter, typically a 0.micron filter. Filter sterilization can be carried out on a high throughput basis if the liposomes have been sized down to about 0.2 to 0.4 microns. Several techniques are available for sizing liposome to a desired size (see, e.g., U.S. Pat. No. 4,737,323).[0180] Suitable lipids include, but are not limited to, DOTMA (Feigner, et al., 1987, Proc. Natl. Acad. Sci. USA 84: 7413-7417), DOGS or Transfectain™ (Behr, et al., 1989, Proc. Natl. Acad. Sci. USA 86: 6982- 6986), DNERIE or DORIE (Feigner, et al., Methods 5: 67-75), DC-CHOL (Gao and Huang, 1991, BBRC 179: 280-285), DOTAPTM (McLachlan, et al., 1995, Gene Therapy 2: 674-622), Lipofectamine™. and glycerolipid compounds (see, e.g., EP901463 and WO98/37916).[0181] Other molecules suitable for complexing with the improved LAMP Constructs include cationic molecules, such as, polyamidoamine (Haensler and Szoka, 1993, Bioconjugate Chern. 4: 372-379), dendritic polysine (WO 95/24221), polyethylene irinine or polypropylene h-nine (WO 96/02655), polylysine (U.S. Pat. No. 5,595,897; FR 2 719 316), chitosan (U.S. Pat. No. 5,744,166), DNA-gelatin coarcervates (see, e.g., U.S. Pat. No. 6,207,195; U.S. Pat. No. 6,025,337; U.S. Pat. No. 5,972,707) or DEAE dextran (Lopata, et al., 1984, Nucleic Acid Res. 12: 5707-5717).
Viral-Based Gene Delivery Vehicles [0182] In one aspect, the improved LAMP Construct delivery vehicle comprises a virus or viral particle. In this aspect, preferably, the improved LAMP Construct comprises a viral vector. Viral vectors, such as retroviruses, adenoviruses, adeno-associated viruses and herpes viruses, are often made up of two WO 2018/204534 PCT/US2018/030725 components, a modified viral genome and a coat structure surrounding it (see, e.g., Smith et al., 1995, Ann. Rev. Microbiol. 49: 807-838), although sometimes viral vectors are introduced in naked form or coated with proteins other than viral proteins. Most current vectors have coat structures similar to a wild-type virus. This structure packages and protects the viral nucleic acid and provides the means to bind and enter target cells.[0183] Preferably, viral vectors comprising the improved LAMP Constructs described herein are modified from wild-type viral genomes to disable the growth of the virus in a target cell while enabling the virus to grow in a host cell (e.g., such as a packaging or helper cell) used to prepare infectious particles. Vector nucleic acids generally essential cis-acting viral sequences for replication and packaging in a helper line and expression control sequences for regulating the expression of a polynucleotide being delivered to a target cell. Other viral functions are expressed in trans in specific packaging or helper cell lines as are known in the art.[0184] Preferred improved LAMP Constructs are viral vectors derived from a virus selected from the group consisting of herpes viruses, cytomegaloviruses, foamy viruses, lentiviruses, Semliki forrest virus, AAV (adeno-associated virus), poxviruses, adenovirases and retroviruses. Such viral vectors are well known in the art.[0185] In one preferred aspect, a viral vector used is an adenoviral vector. The adenoviral genome consists of a linear double-stranded DNA molecule of approximately 36 kb carrying more than about thirty genes necessary to complete the viral replication cycle. The early genes are divided into 4 regions (El to E4) that are essential for viral replication with the exception of the E3 region, which is believed to modulate the anti-viral host immune response. The El region (EIA and EIB) encodes proteins responsible for the regulation of transcription of the viral genome. Expression of the E2 region genes (E2A and E2B) leads to the synthesis of the polypeptides needed for viral replication. The proteins encoded by the E3 region prevent cytolysis by cytotoxic T cells and tumor necrosis factor (Wold and Gooding, 1991, Virology 184: 1-8). The proteins encoded by the E4 region are involved in DNA replication, late gene expression and splicing and host cell shut off (Halbert, et al., 1985, J. Virol. 56: 250-257). The late genes generally encode structural proteins contributing to the viral capsid. In addition, the adenoviral genome carries at cis-acting 5' and 3' ITRs (Inverted Terminal Repeat) and packaging sequences essential for DNA replication. The ITRs harbor origins of DNA replication while the encapsidation region is required for the packaging of adenoviral DNA into infectious particles.[0186] Adenoviral vectors can be engineered to be conditionally replicative (CRAd vectors) in order to replicate selectively in specific cells (e.g., such as proliferative cells) as described in Heise and Kim (2000, J. Clin. Invest. 105: 847-85 1). In another aspect, an adenoviral vector is replication-defective for the El function (e.g., by total or partial deletion or mutagenesis of El). The adenoviral backbone of the vector may comprise additional modifications (deletions, insertions or mutations in one or more viral genes). An example of an E2 modification is illustrated by the thermosensitive mutation localized on the DBF (DNA Binding Protein) encoding gene (Ensinger et al., 1972, J. Virol. 10: 328-339). The adenoviral sequence may also be deleted of all or part of the E4 region (see, e.g., EP 974 668; Christ, et al., 2000, Human Gene Ther. 11: 415- WO 2018/204534 PCT/US2018/030725 427; Lusky, et al., 1999, J. Virol. 73: 8308-8319). Additional deletions within the non-essential E3 region may allow the size of the polynucleotide being delivered to be increased (Yeh, et al., 1997, FASEB Journal 11: 6623). However, it may be advantageous to retain all or part of the E3 sequences coding for polypeptides (e.g., such as gpl9k) allowing the virus to escape the immune system (Gooding, et al., 1990, Critical Review of Immunology 10: 53-71) or inflammatory reactions (EP 00440267.3).[0187] Second generation vectors retaining the ITRs and packaging sequences and comprising substantial genetic modifications to abolish the residual synthesis of the viral antigens also may be used in order to improve long-term expression of the expressed gene in the transduced cells (see, e.g., WO 94/28152; Lusky, et al., 1998, J. Virol 72: 2022-2032).[0188] The improved LAMP Constructs being introduced into the cell may be inserted in any location of the viral genome, with the exception of the cis-acting sequences. Preferably, it is inserted in replacement of a deleted region (El, E3 and/or E4), preferably, within a deleted El region.[0189] Adenoviruses can be derived from any human or animal source, in particular canine (e.g. CAV- or CAV-2 Genbank ref. CAVIGENOM and CAV77082, respectively), avian (Genbank ref. AAVEDSDNA), bovine (such as BAV3; Reddy, et al., 1998, J. Virol. 72: 1394 1402), murine (Genbank ref. ADRMUSMAVI), ovine, feline, porcine or simian sources or alternatively, may be a hybrid virus. Any serotype can be employed. However, the human adenoviruses of the C sub-group are preferred, especially adenoviruses 2 (Ad2) and (Ad5). Such viruses are available, for example, from the ATCC.[0190] Adenoviral particles or empty adenoviral capsids also can be used to transfer improved LAMP Constructs by a virus-mediated cointernalization process as described in U.S. Pat. No. 5,928,944. This process can be accomplished in the presence of cationic agent(s) such as polycarbenes or lipid vesicles comprising one or more lipid layers.[0191] Adenoviral particles may be prepared and propagated according to any conventional technique in the field of the art (e.g., WO 96/17070) using a complementation cell line or a helper virus, which supplies in trans the missing viral genes necessary for viral replication. The cell lines 293 (Graham et al., 1977, J. Gen. Virol. 36: 59-72) and PERC6 (Fallaux et al., 1998, Human Gene Therapy 9: 1909-1917) are commonly used to complement El deletions. Other cell lines have been engineered to complement defective vectors (Yeh, et al., 1996, J. Virol. 70: 559-565; Kroughak and Graham, 1995, Human Gene Ther. 6: 1575-1586; Wang, et al., 1995, Gene Ther. 2: 775-783; Lusky, et al., 1998, J. Virol. 72: 2022-203; EP 919627 and WO 97/04119). The adenoviral particles can be recovered from the culture supernatant but also from the cells after lysis and optionally further purified according to standard techniques (e.g., chromatography, ultracentrifugation, as described in WO 96/27677, WO 98/00524 WO 98/26048 and WO 00/50573).[0192] Cell-type specific targeting may be achieved with vectors derived from adenoviruses having a broad host range by the modification of viral surface proteins. For example, the specificity of infection of adenoviruses is determined by the attachment to cellular receptors present at the surface of permissive cells. In WO 2018/204534 PCT/US2018/030725 this regard, the fiber and penton present at the surface of the adenoviral capsid play a critical role in cellular attachment (Defer, et al., 1990, J. Virol. 64: 3661-3673). Thus, cell targeting of adenoviruses can be carried out by genetic modification of the viral gene encoding fiber and/or penton, to generate modified fiber and/or penton capable of specific interaction with unique cell surface receptors. Examples of such modifications are described in Wickarn, et al., 1997, J. Virol. 71: 8221-8229; Arriberg, et al., 1997, Virol. Chern 268: 6866-6869; Roux, et al., 1989, Proc. Natl. Acad. Sci. USA 86: 9079-9083; Miller and Vile, 1995, FASEB J. 9: 190-199; WO 93/09221, and in WO 95/28494.[0193] In a particularly preferred aspect, adeno-associated viral sequences are used as vectors. Vectors derived from the human parvovirus AAV-2 (adeno-associated virus type 2) are among the most promising gene delivery vehicles currently being developed. Several of the features of this system for packaging a single- stranded DNA suggest it as a possible alternative to naked DNA for delivery. A primary attractive feature, in contrast to other viral vectors such as vaccinia or adenovirus, is that AAV vectors do not express any viral genes. The only viral DNA sequences included in the vaccine construct are the 145 bp inverted terminal repeats (ITR). Thus, as in immunization with naked DNA, the only gene expressed is that of the antigen, or antigen chimera. Additionally, AAV vectors are known to transduce both dividing and non-dividing cells, such as human peripheral blood monocyte-derived dendritic cells, with persistent transgene expression, and with the possibility of oral and intranasal delivery for generation of mucosal immunity. Moreover, the amount of DNA required appears to be much less by several orders of magnitude, with maximum responses at doses of 1010 to 1011 particles or copies of DNA in contrast to naked DNA doses of 50 ug or about 1015 copies.[0194] In one aspect, AAV vectors are packaged by co-transfection of a suitable cell line (e.g., human 293 cells) with the DNA contained in the AAV ITR chimeric protein encoding constructs and an AAV helper plasmid ACG2 containing the AAV coding region (AAV rep and cap genes) without the ITRs. The cells are subsequently infected with the adenovirus Ad5. Vectors can be purified from cell lysates using methods known in the art (e.g., such as cesium chloride density gradient ultracentrifugation) and are validated to ensure that they are free of detectable replication-competent AAV or adenovirus (e.g., by a cytopathic effect bioassay). AAV titer may be determined by quantitative PCR with virus DNA samples prepared after digestion with proteinase K. Preferably, vector titers produced by such a method are approximately 5xl012 to IxlO13 DNase resistant particles per ml.[0195] In other aspects, retroviral vectors are used. Retroviruses are a class of integrative viruses which replicate using a virus-encoded reverse transcriptase, to replicate the viral RNA genome into double stranded DNA which is integrated into chromosomal DNA of the infected cells (e.g., target cells). Such vectors include those derived from murine leukemia viruses, especially Moloney (Gilboa, et al., 1988, Adv. Exp. Med. Biol. 241: 29) or Friend's FB29 strains (WO 95/01447). Generally, a retroviral vector is deleted of all or part of the viral genes gag, pol and env and retains 5' and 3' LTRs and an encapsidation sequence. These elements may be modified to increase expression level or stability of the retroviral vector. Such modifications include the WO 2018/204534 PCT/US2018/030725 replacement of the retroviral encapsidation sequence by one of a retrotransposon such as VL30 (see, e.g., U.S. Pat. No. 5,747,323). Preferably, the improved LAMP Construct is inserted downstream of the encapsidation sequence, preferably in opposite direction relative to the retroviral genome. Cell specific targeting may be achieved by the conjugation of antibodies or antibody fragments to the retroviral envelope protein as is known in the art.[0196] Retroviral particles are prepared in the presence of a helper virus or in an appropriate complementation (packaging) cell line which contains integrated into its genome the retroviral genes for which the retroviral vector is defective (e.g. gag/pol and env). Such cell lines are described in the prior art (Miller and Rosman, 1989, BioTechniques 7: 980; Danos and Mulligan, 1988, Proc. Natl. Acad. Sci. USA 85: 6460; Markowitz, et al., 1988, Virol. 167: 400). The product of the env gene is responsible for the binding of the viral particle to the viral receptors present on the surface of the target cell and, therefore determines the host range of the retroviral particle, in the context of the invention, it is advantageous to use a packaging cell line, such as the PA317 cells (ATCC CRL 9078) or 293EI6 (WO97/35996) containing an amphotropic envelope protein, to allow infection of human and other species' target cells. The retroviral particles are preferably recovered from the culture supernatant and may optionally be further purified according to standard techniques (e.g. chromatography, ultracentrifugation).[0197] Other suitable viruses include poxviruses. The genome of several members of poxyiridae has been mapped and sequenced. A poxyiral vector may be obtained from any member of the poxyiridae, in particular canarypox, fowlpox and vaccinia virus. Suitable vaccinia viruses include, but are not limited to, the Copenhagen strain (Goebel, et al., 1990, Virol. 179: 247-266; Johnson, et al., 1993, Virol. 196: 381-401), the Wyeth strain and the modified Ankara (MVA) strain (Antoine, et al., 1998, Virol. 244: 365-396). The general conditions for constructing a vaccinia virus vector are known in the art (see, e.g., EP 83 286 and EP 206 920; Mayr et al., 1975, Infection 3: 6-14; Sutter and Moss, 1992, Proc. Natl. Acad. Sci. USA 89: 10847-10851). Preferably, the polynucleotide of interest is inserted within a non-essential locus such as the noncoding intergenic regions or any gene for which inactivation or deletion does not significantly impair viral growth and replication.[0198] Poxyiral particles are prepared as described in the art (Piccini, et al., 1987, Methods of Enzymology 153: 545-563; U.S. Pat. No. 4,769,330; U.S. Pat. No. 4,772,848; U.S. Pat. No. 4,603,112; U.S. Pat. No. 5,100,587 and U.S. Pat. No. 5,179,993). Generally, a donor plasmid is constructed, amplified by growth in E. coli and isolated by conventional procedures. Then, it is introduced into a suitable cell culture (e.g. chicken embryo fibroblasts) together with a poxvirus genome, to produce, by homologous recombination, poxyiral particles. These can be recovered from the culture supernatant or from the cultured cells after a lysis step (e.g., chemical lysis, freezing/thawing, osmotic shock, sonication and the like). Consecutive rounds of plaque purification can be used to remove contaminating wild type virus. Viral particles can then be purified using the WO 2018/204534 PCT/US2018/030725 techniques known in the art (e.g., chromatographic methods or ultracentrifugation on cesium chloride or sucrose gradients).[0199] The use of vaccinia as a live virus vaccine in the global campaign to eradicate smallpox made vaccinia an obvious choice for development as a live recombinant vaccine vector. Live recombinant vaccinia viruses expressing close to 100 different foreign proteins have been reported, and a number of these are effective experimental vaccines (reviewed by Moss and Flexner, 1987). Vaccinia is particularly versatile as an expression vector because of its large genomic size, capability of accepting at least 25,000 base pairs of foreign DNA, and its ability to infect most eukaryotic cell types, including insect cells (ibid.). Unlike other DNA viruses, poxviruses replicate exclusively in the cytoplasm of infected cells, reducing the possibility of genetic exchange of recombinant viral DNA with the host chromosome. Recombinant vaccinia vectors have been shown to properly process and express proteins from a variety of sources including man, other mammals, parasites, RNA and DNA viruses, bacteria and bacteriophage.[0200] The expression of DNA encoding a foreign protein is controlled by host virus regulatory elements, including upstream promoter sequences and, where necessary, RNA processing signals. Insertion of foreign DNA into nonessential regions of the vaccinia virus genome has been carried out by homologous recombination (Panicali, et al., Proc. Natl. Acad. Sci, USA, 79: 4927, 1982; Mackett, et al., Proc. Natl. Acad. Sci. USA, 79: 7415, 1982).[0201] Expression of cancer antigens by the improved LAMP Construct may occur because of transcriptional regulatory elements at or near the site of insertion or by more precise genetic engineering. Plasmid vectors that greatly facilitate insertion and expression of foreign genes have been constructed (Mackett, et al., J. Virol, 49: 857, 1982). These vectors contain an expression site, composed of a vaccinia transcriptional promoter and one or more unique restriction endonuclease sites for insertion of the foreign coding sequence flanked by DNA from a nonessential region of the vaccinia genome. The choice of promoter determines both the time (e.g., early or late) and level of expression, whereas the flanking DNA sequence determines the site of homologous recombination.[0202] Only about one in a thousand virus particles produced by this procedure is a recombinant. Although recombinant virus plaques can be identified by DNA hybridization, efficient selection procedures have been developed. By using segments of nonessential vaccinia virus thymidine kinase (TK) gene as flanking sequences, the foreign gene recombines into the TK locus and by insertion inactivates the TK gene. Selection of TK virus is achieved by carrying out the virus plaque assay in TK cells in the presents of 5- bromodeoxyuridine. Phosphorylation of the nucleoside analogue and consequent lethal incorporation into viral DNA occurs only in cells infected with TK+ parental virus. Depending on the efficiency of the transfection and recombination, up to 80 of the plaques are desired recombinants, and the rest are spontaneous TK mutants.[0203] Plasmid vectors that contain the E. colt beta-galactosidase gene, as well as an expression site for a second gene, permit an alternative method of distinguishing recombinant from parental virus (Chakrabarti, WO 2018/204534 PCT/US2018/030725 et al., Mol. Cell. Biol., 5: 3403, 1985). Plaques formed by such recombinants can be positively identified by the blue color that forms upon addition of an appropriate indicator. By combining both TK selection and beta- galactosidase expression, recombinant virus is readily and quickly isolated. The recombinants are then amplified by propagation in suitable cell lines and expression of the inserted gene is checked by appropriate enzymological, immunological or physical procedures.[0204] An upper limit to the amount of genetic information that can be added to the vaccinia virus genome is not yet known. However, the addition of nearly 25,000 base pairs of foreign DNA had no apparent deleterious effect on virus yield (Smith, et al., Gene, 25:21, 1983). Were it necessary, large segments of the vaccinia virus genome could be deleted to provide additional capacity (Moss, et al., J. Virol. 40: 387, 1981).[0205] Viral capsid molecules may include targeting moieties to facilitate targeting and/or entry into cells. Suitable targeting molecules, include, but are not limited to: chemical conjugates, lipids, glycolipids, hormones, sugars, polymers (e.g. PEG, polylysine, PEI and the like), peptides, polypeptides (see, e.g., WO 94/40958), vitamins, antigens, lectins, antibodies and fragments thereof. Preferably, such targeting molecules recognize and bind to cell-specific markers, tissue-specific markers, cellular receptors, viral antigens, antigenic epitopes or tumor-associated markers.[0206] Compositions comprising an improved LAMP Construct based on viral particles may be formulated in the form of doses of between 10 and 1014 i.u. (infectious units), and preferably, between 10 and 1011 i.u. The titer may be determined by conventional techniques. The doses of LAMP Constructs are preferably comprised between 0.01 and 10 mg/kg, more especially between 0.1 and 2 mg/kg.
Self-Replicating RNA [0207] Self-replicating RNA virus vectors can also be constructed using the improved LAMP Constructs as described herein. For example, alphaviruses, flavivuses, measle virus and rhabdoviruses can be used to generate self-replicating RNA virus vaccines. Preferred strains of self-replicating RNA viruses include, but are not limited to rabies virus (RABV), vesicular stomatisitis virus (VSV), West Nile virus, Kunjin virus, Semliki Forest virus (SFV), Sindbis virus (SIN) and/or Venezuelan equine encephalitis virus (VEE).[0208] Self-replicating RNA viruses express the native antigen upon delivery into tissue, thus mimicking live attenuated vaccines without having the risk of reversion to pathogenicity. They also stimulate the innate immune system, thus potentiating responses. See, e.g., Ljungberg, K. "Self-replicating alphavirus RNA vaccines," Expert Rev Vaccines (2): 177-94 (2015); Lundstrom, K., "Oncolytic Alphaviruses in Cancer Immunotherapy", Vaccines 5:9 (2017); Lundstrom, K. "Replicon RNA Viral Vectors as Vaccines," Vaccines 4:39 (2016) (hereby incorporated by reference in their entirety). Use of self-replicating vaccines comprising the improved LAMP Constructs described herein can also be used in prime-boost protocols.[0209] Moreover, self-replicating RNA viruses can also be encapsulated by liposomes, as described herein, to improve delivery and targeting. Immunization with self-replicating RNA viruses comprising the WO 2018/204534 PCT/US2018/030725 improved LAMP Constructs described herein may provide higher transient expression levels of antigens resulting in generation of neutralizing antibody responses and protection against lethal challenges under safe conditions.
Cell-Based Delivery Vehicles [0210] The improved LAMP Constructs according to the invention can be delivered to target cells by means of other cells ("delivery cells") which comprise the constructs. Methods for introducing constructs into cells are known in the art and include microinjection of DNA into the nucleus of a cell (Capechi, et al., 1980, Cell 22: 479-488); transfection with CaP04 (Chen and Okayama, 1987, Mol. Cell Biol. 7: 2745 2752), electroporation (Chu, et al., 1987, Nucleic Acid Res. 15: 1311-1326); lipofection/liposome fusion (Feigner, et al., 1987, Proc. Natl. Acad. Sci. USA 84: 7413-7417) and particle bombardment (Yang, et al., 1990, Proc. Natl. Acad. Sci. USA 87: 9568-9572). Suitable cells include autologous and non-autologous cells, and may include xenogenic cells. Delivery cells may be induced to deliver their contents to the target cells by inducing their death (e.g., by providing inducible suicide genes to these cells).
Accessory Molecules [0211] The compositions comprising the improved LAMP Constructs according to the invention may comprise one or more accessory molecules for facilitating the introduction of an improved LAMP Construct into a cell and/or for enhancing a particular therapeutic effect and/or enhancing antibody production.[0212] In addition, the composition comprising the improved LAMP Construct according to the present invention may include one or more stabilizing substance(s), such as lipids, nuclease inhibitors, hydrogels, hyaluronidase (WO 98/53853), collagenase, polymers, chelating agents (EP 890362), in order to inhibit degradation within the animal/human body and/or improve transfection/infection of the vector into a target cell. Such substances may be used alone or in combination (e.g., cationic and neutral lipids).[0213] It has also been shown that adenovirus proteins are capable of destabilizing endosomes and enhancing the uptake of DNA into cells. The mixture of adenoviruses to solutions containing a lipid-complexed DNA vector or the binding of DNA to polylysine covalently attached to adenoviruses using protein cross- linking agents may substantially improve the uptake and expression of an improved LAMP Construct (see, e.g., Curiel, et al., 1992, Am. I. Respir. Cell. Mol. Biol. 6: 247-252).
Host Cells [0214] Improved LAMP Constructs according to the invention can be expressed in a variety of host cells, including, but not limited to: prokaryotic cells (e.g., E. coli, Staphylococcus sp., Bacillus sp.); yeast cells (e.g., Saccharomyces sp.); insect cells; nematode cells; plant cells; amphibian cells (e.g., Xenopus); avian cells; WO 2018/204534 PCT/US2018/030725 and mammalian cells (e.g., human cells, mouse cells, mammalian cell lines, primary cultured mammalian cells, such as from dissected tissues).[0215] The molecules can be expressed in host cells isolated from an organism, host cells which are part of an organism, or host cells which are introduced into an organism. In one aspect, improved LAMP Constructs are expressed in host cells in vitro, e.g., in culture. In another aspect, improved LAMP Constructs are expressed in a transgenic organism (e.g., a transgenic mouse, rat, rabbit, pig, primate, etc.) that comprises somatic and/or germline cells comprising nucleic acids encoding the improved LAMP Constructs. Methods for constructing transgenic animals are well known in the art and are routine.[0216] Improved LAMP Constructs also can be introduced into cells in vitro, and the cells (e.g., such as stem cells, hematopoietic cells, lymphocytes, and the like) can be introduced into the host organism. The cells may be heterologous or autologous with respect to the host organism. For example, cells can be obtained from the host organism, improved LAMP Constructs introduced into the cells in vitro, and then reintroduced into the host organism.
Antigen Presenting Cells [0217] In a preferred aspect of the invention, an improved LAMP Construct as described herein is introduced into a natural or engineered antigen presenting cell.[0218] The term "antigen presenting cell" (APC) as used herein intends any cell which presents on its surface an antigen in association with a major histocompatibility complex molecule, preferably a class II molecule, or portion thereof. Examples of suitable APCs are discussed in detail below and include, but are not limited to, whole cells such as macrophages, dendritic cells, B cells, hybrid APCs, and foster antigen presenting cells. Methods of making hybrid APCs are described and known in the art.[0219] Dendritic cells (DCs) are potent antigen-presenting cells. It has been shown that DCs provide all the signals required for T cell activation and proliferation. These signals can be categorized into two types. The first type, which gives specificity to the immune response, is mediated through interaction between the T- cell receptor/CD3 ("TCR/CD3") complex and an antigenic peptide presented by a major histocompatibility complex ("MHC" defined above) class I or II protein on the surface of APCs. This interaction is necessary, but not sufficient, for T cell activation to occur. In fact, without the second type of signals, the first type of signals can result in T cell anergy. The second type of signals, called co-stimulatory signals, is neither antigen-specific nor MHC-restricted, and can lead to a full proliferation response of T cells and induction of T cell effector functions in the presence of the first type of signals.[0220] Several molecules have been shown to enhance co-stimulatory activity. These include, but are not limited to, heat stable antigen (HSA), chondroitin sulfate-modified MHC invariant chain (li-CS), intracellular adhesion molecule I (ICAM-1), and B7 co-stimulatory molecule on the surface of APCs and its counter-receptor CD28 or CTLA-4 on T cells.
WO 2018/204534 PCT/US2018/030725 id="p-221" id="p-221" id="p-221"
[0221] Other important co-stimulatory molecules are CD40, CD54, CD80, CD86. As used herein, the term "co-stimulatory molecule" encompasses any single molecule or combination of molecules which, when acting together with a peptide/MHC complex bound by a TCR on the surface of a T cell, provides a co- stimulatory effect which achieves activation of the T cell that binds the peptide. The term thus encompasses B7, or other co-stimulatory molecule(s) on an APC, fragments thereof (alone, complexed with another molecule(s), or as part of a fusion protein) which, together with peptide/MHC complex, binds to a cognate ligand and result in activation of the T cell when the TCR on the surface of the T cell specifically binds the peptide. Co-stimulatory molecules are commercially available from a variety of sources, including, for example, Beckman Coulter.[0222] In one aspect of the invention, the method described in Romani et al., J. Immunol. Methods 196: 135-151, 1996, and Bender et al, J. Immunol. Methods 196: 121-135, 1996, are used to generate both immature and mature dendritic cells from the peripheral blood mononuclear cells (PBMCs) of a mammal, such as a murine, simian or human. Briefly, isolated PBMCs are pre-treated to deplete T- and B-cells by means of an immunomagnetic technique. Lymphocyte-depleted PBMC are then cultured for in RPMI medium 9 e.g., about 7 days), supplemented with human plasma (preferably autologous plasma) and GM-CSF/IL-4, to generate dendritic cells. Dendritic cells are nonadherent when compared to their monocyte progenitors. Thus, on approximately day 7, non-adherent cells are harvested for further processing.[0223] The dendritic cells derived from PBMC in the presence of GM-CSF and IL-4 are immature, in that they can lost the nonadherence property and revert back to macrophage cell fate if the cytokine stimuli are removed from the culture. The dendritic cells in an immature state are very effective in processing native protein antigens for the MHC class II restricted pathway (Romani, et al., J. Exp. Med. 169:1169, 1989). Further maturation of cultured dendritic cells is accomplished by culturing for 3 days in a macrophage-conditioned medium (CM), which contains the necessary maturation factors. Mature dendritic cells are less able to capture new proteins for presentation but are much better at stimulating resting T cells (both CD4 and CDS) to grow and differentiate.[0224] Mature dendritic cells can be identified by their change in morphology, such as the formation of more motile cytoplasmic processes; by their nonadherence; by the presence of at least one of the following markers: CD83, CD68, HLA-DRor CD86; or by the loss of Fc receptors such as CD 115 (reviewed in Steinman, Annu. Rev. Immunol. 9: 271, 1991). Mature dendritic cells can be collected and analyzed using typical cytofluorography and cell sorting techniques and devices, such as FACScan and FACStar. Primary antibodies used for flow cytometry are those specific to cell surface antigens of mature dendritic cells and are commercially available. Secondary antibodies can be biotinylated Igs followed by FITC- or PE-conjugated streptavidin.[0225] Alternatively, others have reported that a method for upregulating (activating) dendritic cells and converting monocytes to an activated dendritic cell phenotype. This method involves the addition of calcium ionophore to the culture media convert monocytes into activated dendritic cells. Adding the calcium WO 2018/204534 PCT/US2018/030725 21 ionophore A23187, for example, at the beginning of a 24-48 hour culture period resulted in uniform activation and dendritic cell phenotypic conversion of the pooled "monocyte plus DC" fractions: characteristically, the activated population becomes uniformly CD 14 (Leu M3) negative, and upregulates HLA-DR, HLA-DQ, ICAM-1,137.1, and 137.2. Furthermore, this activated bulk population functions as well on a small numbers basis as a further purified. Specific combination(s) of cytokines have been used successfully to amplify (or partially substitute) for the activation/conversion achieved with calcium ionophore: these cytokines include but are not limited to G-CSF, GM-CSF, IL-2, and IL-4. Each cytokine when given alone is inadequate for optimal upregulation.[0226] The second approach for isolating APCs is to collect the relatively large numbers of precommitted APCs already circulating in the blood. Previous techniques for isolating committed APCs from human peripheral blood have involved combinations of physical procedures such as metrizamide gradients and adherence/nonadherence steps (Freudenthal et al. PNAS 87: 7698-7702, 1990); Percoll gradient separations (Mehta-Damani, et al., J. Immunol. 153: 996-1003, 1994); and fluorescence activated cell sorting techniques (Thomas et al., J. Immunol. 151: 6840-52, 1993).[0227] There are many other methods routine in the art for isolating professional antigen presenting cells (or their precursors) and that such methods and others which may be developed are not limiting and are encompassed within the scope of the invention.[0228] In one embodiment, the APCs and therefore the cells presenting one or more antigens are autologous. In another embodiment, the APCs presenting the antigen are allogeneic, i.e., derived from a different subject.[0229] As discussed herein, improved LAMP Constructs can be introduced into APCs using the methods described above or others known in the art, including, but not limited to, transfection, electroporation, fusion, microinjection, viral-based delivery, or cell based delivery. Arthur et al., Cancer Gene Therapy 4(1): 17- 25, 1997, reports a comparison of gene transfer methods in human dendritic cells.[0230] Known, partial and putative human leukocyte antigen (HLA), the genetic designation for the human MHC, amino acid and nucleotide sequences, including the consensus sequence, are published (see, e.g., Zemmour and Parham, Immunogenetics 33: 310-320, 1991), and cell lines expressing HLA variants are known and generally available as well, many from the American Type Culture Collection ("ATCC"). Therefore, using PCR, MHC class Il-encoding nucleotide sequences are readily operatively linked to an expression vector of this invention that is then used to transform an appropriate cell for expression therein.[0231] Professional APCs can be used, such as macrophages, B cells, monocytes, dendritic cells, and Langerhans cells. These are collected from the blood or tissue of 1) an autologous donor; 2) a heterologous donor having a different HLA specificity then the host to be treated; or 3) from a xenogeneic donor of a different species using standard procedures (Coligan, et. al., Current Protocols in Immunology, sections 3 and 14, 1994).
WO 2018/204534 PCT/US2018/030725 The cells may be isolated from a normal host or a patient having an infectious disease, cancer, autoimmune disease, or allergy.[0232] Professional APCs may be obtained from the peripheral blood using leukopheresis and "FICOLL/HYPAQUE" density gradient centrifugation (stepwise centrifugation through Ficoll and discontinuous Percoll density gradients). Procedures are utilized which avoid the exposure of the APCs to antigens which could be internalized by the APCs, leading to activation of T cells not specific for the antigens of interest.[0233] Cells which are not naturally antigen presenting can be engineered to be antigen presenting by introducing sequences encoding appropriate molecules. For example, nucleic acid sequences encoding MHC class II molecules, accessory molecules, co-stimulatory molecules and antigen processing assisting molecules can be introduced after direct synthesis, cloning, purification of DNA from cells containing such genes, and the like. One expedient means to obtain genes for encoding the molecules used in the improved LAMP Constructs and methods described herein is by polymerase chain reaction (PCR) amplification on selected nucleic acid templates with selected oligonucleotide primer pairs. For example, epithelial cells, endothelial cells, tumor cells, fibroblasts, activated T cells, eosinophils, keratinocytes, astrocytes, microglial cells, thymic cortical epithelial cells, Schwann cells, retinal pigment epithelial cells, myoblasts, vascular smooth muscle cells, chondrocytes, enterocytes, thyrocytes and kidney tubule cells can be used. These may be primary cells recently explanted from a host and not extensively passaged in cell culture to form an established cell line, or established cell lines that are relatively homogeneous and capable of proliferating for many generations or indefinitely.[0234] Cells that are not professional APCs are isolated from any tissue of an autologous donor; a heterologous donor or a xenogeneic donor, where they reside using a variety of known separation methods (Darling, Animal Cells: Culture and Media. J. Wiley, New York, 1994; Freshney, Culture of Animal Cells. Alan R. Liss, Inc., New York, 1987). Non-autologous cells, e.g., heterologous or xenogeneic cells, can be engineered ex vivo to express HLA class I and class II molecules that match known human HLA specificities. These cells can then be introduced into a human subject matching the HLA specificity of the engineered cells. The cells are further engineered ex vivo to express one or more LAMP Constructs according to the invention.[0235] The engineered cells are maintained in cell culture by standard cell culture methods (Darling, Animal Cells: Culture and Media". J. Wiley, New York, 1994; Freshney, Culture of Animal Cells". Alan R. Liss, Inc., New York, 1987). Cell lines for use in the present invention are obtained from a variety of sources (e.g., ATCC Catalogue of Cell Lines & Hybidomas, American Type Culture Collection, Sth edition, 1995), or are produced using standard methods (Freshney, Culture of Immortalized Cells, Wiley-Liss, New York, 1996). Non-transformed cell lines are preferred for use in human subjects.[0236] In one aspect, CD34+ precursors that are differentiating under the influence of GM-CSF into dendritic cells are obtained from the body of a subject and nucleic acids encoding LAMP Constructs according to the invention are introduced into the cells, which are then injected into the subject. Utilizing the improved WO 2018/204534 PCT/US2018/030725 LAMP Constructs as described herein will enhance the association of peptides derived from a particular antigen with MHC class II molecules on the transduced antigen presenting cells, resulting in significantly more potent systemic T cell dependent immune responses and/or antibody production. While the antigen presenting cells transfected in this strategy are preferably autologous cells, any MHC class II cells that effectively present antigen in the host may be used as described above.
Peptide Vaccines [0237] Also within the scope of this invention are peptide vaccines encoded by the improved LAMP Construct Preferably, the antigen is processed within the compartment/organelle (or subsequent compartment/organelle to which it is delivered) to generate an epitope bound to an MHC class II molecule capable of modulating an immune response.[0238] The peptide vaccines encoded by the improved LAMP Constructs may also may be bound in a membranous structure to facilitate its administration to the body of an organism. For example, the peptide vaccine encoded by the improved LAMP Construct may be incorporated into liposomes, as described in U.S. Pat. No. 4,448,765.[0239] When a protein or polypeptide is to be used as an immunogen, it may be produced by expression of any one or more of the improved LAMP Constructs described herein in a recombinant cell or it may be prepared by chemical synthesis. For example, the Merrifield technique (Journal of American Chemical Society, vol. 85, pp. 2149-2154, 1968), can be used.
Methods of Producing Antibodies using LAMP Constructs [0240] The improved LAMP Constructs as polynucleotides, the encoded proteins of the improved LAMP Constructs, and/or cells (such as antigen presenting cells which express the improved LAMP Constructs described herein) can be used to generate antibodies by methods well known by the skilled artisan, such as, for example, methods described in the art. See, for instance, Sutcliffe et al., supra; Wilson et al., supra; Chow et al., Proc. Natl. Acad. Sci. USA 82:910-914 (1985); and Bittie et al., J. Gen. Virol. 66:2347-2354 (1985). If in vivo immunization is used, animals may be immunized with a protein encoded by the improved LAMP Construct and/or a polynucleotide comprising the improved LAMP Construct comprising an antigen as described herein. Priming with improved LAMP Constructs as polynucleotides, the encoded proteins of the improved LAMP Constructs, and/or cells (such as antigen presenting cells which express the improved LAMP Constructs described herein) followed by boosting with an antigen is a preferred embodiment of the invention. In further preferred embodiments, priming with an improved LAMP Construct as described herein followed by boosting with an antigen is specifically contemplated and can be used to generate an even more robust immune response, especially in view of antibody repertoire diversity and titer.
WO 2018/204534 PCT/US2018/030725 id="p-241" id="p-241" id="p-241"
[0241] The improved LAMP Construct comprising the antigen may be injected into the non-human vertebrate to raise antibodies. Preparation and injection of LAMP Constructs into non-human vertebrates can be accomplished according to principles of immunization of animals that are well known to those skilled in the art.[0242] The use of an improved LAMP Construct to effectively present the antigen involves, in one aspect, the antigen being processed by LAMP in Antigen Presenting Cells after endocytosis and fusion of the endosome with a lysosome. The endosome then merges with an exocytic vesicle from the Golgi apparatus containing class II MHC molecules, to which the resultant peptides bind. The MHC-peptide complex then trafficks to the plasma membrane where the antigen is available for display to CD4+ T cells.[0243] Animals such as rabbits, rats, mice, llamas, camels, and/or cows can be immunized with the improved LAMP Construct comprising an antigen and/or a polynucleotide encoding the improved LAMP Construct comprising an antigen. Additional animals suitable for immunization include, non-human mammals, such as a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon, rhesus macaque), an ape (e.g. gorilla, chimpanzee, orangutan, gibbon).[0244] For instance, intraperitoneal and/or intradermal injection of emulsions containing about 1micrograms of an improved LAMP Construct comprising an antigen or carrier protein and Freund's adjuvant or any other adjuvant known for stimulating an immune response may be used. Several booster injections (such as with the recombinant antigen protein) may be needed, for instance, at intervals of about two weeks, to provide a useful titer of an anti-antigen antibody which can be detected, for example, by ELISA assay using free antigen adsorbed, directly or indirectly (e.g., via a biotinylated AviTag), to a solid surface. The titer of anti-antigen antibodies in serum from an immunized animal may be increased by selection of anti-antibodies, for instance, by adsorption to the antigen on a solid support and elution of the selected antibodies according to methods well known in the art.[0245] Alternatively, a polynucleotide encoding the improved LAMP Construct comprising an antigen can also be directly introduced into animals. See, for example, U.S. patent numbers 5,676,954; 6,875,748; 5,661,133; Sahin et al., Nat Rev Drug Discov, 2014 Oct;13(10):759-80; Kariko et al., Mol Ther, 20Nov;16(ll):1833-40; Kariko et al., Nucleic Acid Res, 2011, Nov;39(21):el42; US Patent number 6,511,832. In one example, an improved LAMP Construct comprising an antigen is directly injected into a non-human vertebrate. Injection into the animals can occur via intramuscular, intradermal, intranasal, subcutaneous, intravenous, intratracheal, and intrathecal deliveries. Follow-on boosting with a recombinant antigen can also be include in generating the antibodies.[0246] Additionally, antibodies generated by the disclosed methods can be affinity matured using display technology, such as for example, phage display, yeast display or ribosome display. In one example, single chain antibody molecules ("scFvs") displayed on the surface of phage particles are screened to identify WO 2018/204534 PCT/US2018/030725 those scFvs that immunospecifically bind to the antigen and/or the starting protein. The present invention encompasses both scFvs and portions thereof that are identified to immunospecifically bind to the antigen and/or the starting protein. Such scFvs can routinely be "converted" to immunoglobulin molecules by inserting, for example, the nucleotide sequences encoding the VH and/or VL domains of the scFv into an expression vector containing the constant domain sequences and engineered to direct the expression of the immunoglobulin molecule.[0247] Recombinant expression of the raised antibodies (including scFvs and other molecules comprising, or alternatively consisting of, antibody fragments or variants thereof (e.g., a heavy or light chain of an antibody of the invention or a portion thereof or a single chain antibody of the invention)) using the improved LAMP Construct comprising an antigen and/or a polynucleotide encoding the improved LAMP Construct comprising an antigen of the invention, requires construction of an expression vector(s) containing a polynucleotide that encodes the antibody or fragment or variant thereof. Once a polynucleotide encoding an antibody molecule (e.g., a whole antibody, a heavy or light chain of an antibody, or variant or portion thereof (preferably, but not necessarily, containing the heavy or light chain variable domain)), of the invention has been obtained, the vector(s) for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing an antibody by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination and are described herein. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding the anti- antigen antibody obtained and isolated as described herein (e.g., a whole antibody, a heavy or light chain of an antibody, a heavy or light chain variable domain of an antibody, or a portion thereof, or a heavy or light chain CDR, a single chain Fv, or fragments or variants thereof), operably linked to a promoter. Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Patent No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy chain, the entire light chain, or both the entire heavy and light chains.[0248] The expression vector(s) can be transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce either the anti-antigen antibody. Thus, the invention includes host cells containing polynucleotide(s) encoding the anti-antigen antibody (e.g., whole antibody, a heavy or light chain thereof, or portion thereof, or a single chain antibody of the invention, or a fragment or variant thereof), operably linked to a heterologous promoter. In preferred embodiments, for the expression of entire antibody molecules, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
WO 2018/204534 PCT/US2018/030725 id="p-249" id="p-249" id="p-249"
[0249] A variety of host-expression vector systems may be utilized to express anti-antigen antibody. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected, with the appropriate nucleotide coding sequences, express the anti-antigen antibody. These include, but are not limited to, microorganisms such as bacteria (e.g., E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, are used for the expression of the anti-antigen antibody. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).[0250] In bacterial systems, a number of expression vectors may be advantageously selected depending upon the intended use. For example, when a large quantity of a protein is to be produced (for either antibody production or encoded polypeptides of the improved LAMP Construct), vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., EMBO 1. 2:1791 (1983)), in which the coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chern. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione 5-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or Factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.[0251] In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) may be used as a vector to express an anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct. The virus grows in Spodoptera frugiperda cells. Coding sequences may be cloned individually into non­ WO 2018/204534 PCT/US2018/030725 essential regions (for example, the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example, the polyhedrin promoter).[0252] In mammalian host cells, a number of viral-based expression systems may be utilized express an anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct. In cases where an adenovirus is used as an expression vector, the coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.[0253] Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct in infected hosts (e.g., see Logan & Shenk, Proc. Natl. Acad. Sci. USA 1:355-359 (1984)).[0254] Specific initiation signals may also be required for efficient translation of inserted coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see, e.g., Bittner et al., Methods in Enzymol. 153:51-544(1987)).[0255] In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed, to this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include, but are not limited to, CHO, VERY, BHK, Hela, COS, NSO, MDCK, 293, 3T3, W138, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT2O and T47D, and normal mammary gland cell line such as, for example, CRL7O3O and HsS78Bst.[0256] For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the express an anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with a polynucleotide controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign polynucleotide, engineered cells may be WO 2018/204534 PCT/US2018/030725 allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct.[0257] A number of selection systems may be used, including but not limited to, the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)), hypoxanthineguanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:8 17 (1980)) genes can be employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 (Goldspiel et al., Clinical Pharmacy, 12: 488-505 (1993); Wu and Wu, Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62: 191-217 (1993); TIB TECH 11(5): 155-2 15 (May; 1993)); and hygro, which confers resistance to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example; in Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993); Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990); and in Chapters 12 and 13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John Wiley & Sons, NY (1994); Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981).[0258] The expression levels of either an anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct can be increased by vector amplification (for a review, see Bebbington and Hentschel, The Use Of Vectors Based On Gene Amplification For The Expression Of Cloned Genes In Mammalian Cells In DNA Cloning, Vol.3. (Academic Press, New York, 1987)). When a marker in the vector system expressing an anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct is amplifiable, an increase in the level of inhibitor present in the host cell culture will increase the number of copies of the marker gene. Since the amplified region is associated with the coding sequence, production of the anti-antigen antibody express or the encoded polypeptides of the improved LAMP Construct will also increase (Crouse et al., Mol. Cell. Biol. 3:257 (1983)).[0259] Other elements that can be included in vector sequences include heterologous signal peptides (secretion signals), membrane anchoring sequences, introns, alternative splice sites, translation start and stop signals, inteins, biotinylation sites and other sites promoting post-translational modifications, purification tags, sequences encoding fusions to other proteins or peptides, separate coding regions separated by internal WO 2018/204534 PCT/US2018/030725 ribosome reentry sites, sequences encoding "marker" proteins that, for example, confer selectability (e.g., antibiotic resistance) or sortability (e.g., fluorescence), modified nucleotides, and other known polynucleotide cis-acting features not limited to these examples.[0260] The host cell may be co-transfected with two expression vectors of the invention, for example, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain is preferably placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2 197 (1980)). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA or synthetic DNA sequences.[0261] Once an anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct has been produced by recombinant expression, it may be purified by any method known in the art for purification of a protein, for example, by chromatography (e.g., ion exchange, affinity (particularly by Protein A affinity and immunoaffinity for the specific antigen), and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. Further, an anti- antigen antibody or the encoded polypeptides of the improved LAMP Construct may be fused to heterologous polypeptide sequences described herein or otherwise known in the art to facilitate purification.[0262] In one example, the anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any combination thereof and portions thereof), or albumin (including but not limited to recombinant human albumin or fragments or variants thereof (see, e.g., U.S. Patent No. 5,876,969, issued March 2,1999, EP Patent 0 413 622, and U.S. Patent No. 5,766,883, issued June 16,1998), resulting in chimeric polypeptides. Such fusion proteins may facilitate purification and may increase half-life in vivo. This has been shown for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins. See, e.g., EP 394,827; Traunecker et al., Nature, 331:84-86 (1988). Enhanced delivery of an antigen across the epithelial barrier to the immune system has been demonstrated for antigens (e.g., insulin) conjugated to an FcRn binding partner such as IgG or Fe fragments (see, e.g., PCT Publications WO 96/22024 and WO 99/04813). IgG Fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion disulfide bonds have also been found to be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone. See, e.g., Fountoulakis et al., J. Biochem., 270:3958-3964 (1995). Nucleic acids encoding the anti-antigen antibody or the encoded polypeptides of the improved LAMP Construct described herein can also be recombined with a gene of interest as an epitope tag (e.g., the hemagglutinin ("HA") tag or flag tag) to aid in detection and purification of the expressed polypeptide. For example, a system described by WO 2018/204534 PCT/US2018/030725 Janknecht et al. allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad. Sci. USA 88:8972- 897). In this system, the gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the gene is translationally fused to an amino-terminal tag consisting of six histidine residues. The tag serves as a matrix-binding domain for the fusion protein. Extracts from cells infected with the recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose column and histidine-tagged proteins can be selectively eluted with imidazole- containing buffers.
Administration [0263] Vaccine material according to this invention may contain the immune stimulatory improved LAMP Constructs described herein or may be recombinant microorganisms, or antigen presenting cells which express the immune stimulatory improved LAMP Constructs. Preparation of improved LAMP Constructs containing vaccine material according to this invention and administration of such improved LAMP Constructs for immunization of individuals are accomplished according to principles of immunization that are well known to those skilled in the art.[0264] Large quantities of these materials may be obtained by culturing recombinant or transformed cells containing replicons that express the improved LAMP Constructs described herein. Culturing methods are well-known to those skilled in the art and are taught in one or more of the documents cited above. The improved LAMP Construct vaccines are generally produced by culture of recombinant or transformed cells and formulated in a pharmacologically acceptable solution or suspension, which is usually a physiologically- compatible aqueous solution, or in coated tablets, tablets, capsules, suppositories or ampules, as described in the art, for example in U.S. Pat. No. 4,446,128, incorporated herein by reference. Administration may be any suitable route, including oral, rectal, intranasal or by injection where injection may be, for example, transdermal, subcutaneous, intramuscular or intravenous.[0265] The improved LAMP Constructs are administered to a mammal in an amount sufficient to induce an immune response in the mammal. A minimum preferred amount for administration is the amount required to elicit antibody formation to a concentration at least 4 times that which existed prior to administration. A typical initial dose for administration would be 10-5000 micrograms when administered intravenously, intramuscularly or subcutaneously, or 105 to 1011 plaque forming units of a recombinant vector, although this amount may be adjusted by a clinician doing the administration as commonly occurs in the administration of vaccines and other agents which induce immune responses. A single administration may usually be sufficient to induce immunity, but multiple administrations may be carried out to assure or boost the response.[0266] The improved LAMP Construct vaccines may be tested initially in a non-human mammal (e.g., a mouse or primate). For example, assays of the immune responses of inoculated mice can be used to demonstrate greater antibody, T cell proliferation, and cytotoxic T cell responses to the improved LAMP WO 2018/204534 PCT/US2018/030725 Constructs than to wild type antigen. Improved LAMP Constructs can be evaluated in Rhesus monkeys to determine whether the vaccine formulation that is highly effective in mice will also elicit an appropriate monkey immune response. In one aspect, each monkey receives a total of 5 mg DNA per immunization, delivered IM and divided between 2 sites, with immunizations at day 0 and at weeks 4, 8, and 20, with additional doses optional. Antibody responses, ADCC, CD4+ and CD8+ T-cell cytokine production, CD4+ and CD8+ T-cell antigen-specific cytokine staining can be measured to monitor immune responses to the vaccine.[0267] Further description of suitable methods of formulation and administration according to this invention may be found in U.S. Pat. No. 4,454,116 (constructs), U.S. Pat. No. 4,681,762 (recombinant bacteria), and U.S. Pat. 4,592,002 and 4,920,209 (recombinant viruses).
Cancer Immunotherapy: Candidates for Prevention and Treatment [0268] The improved LAMP Constructs described herein comprising at least one cancer antigen described in Table 1 can be used to treat patients who have cancer and/or a hyperproliferative disorder. Examples include patients with documented Epstein-Barr virus associated lymphomas, patients with HPV associated cervical carcinomas, patients with chronic HCV, or patients with a defined re-arrangement or mutation in an oncogene or tumor suppressor gene. Further preferred embodiments include patients having hyperproliferative disorders, including cancer, resulting from CMV infection.[0269] In preferred embodiments, cancers that can be treated using the improved LAMP Constructs described herein include, but are not limited to all stages of progression, including hyperplasia of an adenocarcinoma, sarcoma, skin cancer, melanoma, bladder cancer, brain cancer (including glioblastoma multiforme), breast cancer, uterine cancer, ovarian cancer, prostate cancer, lung cancer (including, but not limited to NSCLC, SCLC, squamous cell cancer), colorectal cancer, anal cancer, rectal cancer, cervical cancer, liver cancer, head and neck cancer, oral cancer, salivary gland cancer, esophageal cancer, pancreas cancer, pancreatic ductal adenocarcinoma (PDA), renal cancer, stomach cancer, kidney cancer, multiple myeloma or cerebral cancer.[0270] It is envisioned that therapy with an improved LAMP Construct, such as in the case of a vaccine composition comprising the improved LAMP Construct, could be utilized at any period during the course of the individual's cancer, once it is identified. It is also possible that in high risk patients, vaccination in order to prevent the subsequent emergence of a cancer. Protein based LAMP Construct vaccines as well as cell therapy (e.g, dendritic cell therapy comprising LAMP Constructs) are also envisioned.
Procedure for Therapy [0271] In one embodiment, the improved LAMP Constructs could be injected into the patient at any suitable time during the course of their malignancy. For example, the improved LAMP Construct comprising at least one cancer antigen described in Table 1 would be injected at a stage when the tumor burden was low.
WO 2018/204534 PCT/US2018/030725 In an alternative embodiment in which the improved LAMP Construct is introduced into the individual's antigen presenting cells (such as, for example, dendritic cells), precursors to the antigen presenting cells or mature antigen presenting cells are drawn either from the individual's bone marrow or peripheral blood by vena puncture. These cells are established in culture followed by transduction with the improved LAMP Construct. Once transduction had occurred, these antigen presenting cells are injected back into the patient.[0272] In a particularly preferred embodiment, the invention provides a method of treatment for a cancer patient having low tumor burden, such as early in the disease, after resection of a neoplastic tumor, or when the burden of tumor cells is otherwise reduced. In this method, a cell population containing autologous stem cells capable of differentiation into antigen presenting cells which will express MHC class II molecules is obtained from the patient. These cells are cultured and transformed by introducing an improved LAMP Construct to deliver the cancer antigen to be associated with an MHC class II molecule either within the compartment/organelle or within another compartment/organelle to which the antigen is delivered.[0273] The transfected stem cell population is then reintroduced into the patient, where the stem cells differentiate into antigen presenting cells which express MHC class II molecules complexed with Th epitopes from the antigen. The immune response to the antigen will be enhanced by enhanced stimulation of the helper T cell population.[0274] More generally, in one embodiment, this invention provides a vaccine composition comprising the improved LAMP Construct for modulating an immune response in a mammal to an antigen (i.e., stimulating, enhancing, or reducing such a response).
Kits [0275] The invention further comprises kits to facilitate performing the methods described herein. In one aspect, a kit comprises an improved LAMP Construct as described herein and a cell for receiving the improved LAMP Construct. The kit may additionally comprise one or more nucleic acids for engineering the cell into a professional APC. In one aspect, however, the cell is a professional APC. The cell may or may not express co-stimulatory molecules. In a preferred aspect, when the cell does not express co-stimulatory molecules, the antigen encoded by the improved LAMP Construct is an autoantigen. In another aspect, a panel of cells is provided expressing different MHC molecules (e.g., known to be expressed in human beings). In a further aspect, the kit comprises reagents to facilitate entry of the improved LAMP Constructs into a cell (e.g., lipid-based formulations, viral packaging materials, cells, and the like). In still a further aspect, one or more T cell lines specific for the antigen encoded by the improved LAMP Construct is provided, to verify the ability of the improved LAMP Construct to elicit, modulate, or enhance an immune response.
EXAMPLES WO 2018/204534 PCT/US2018/030725 id="p-276" id="p-276" id="p-276"
[0276] The invention will now be further illustrated with reference to the following examples. It will be appreciated that what follows is by way of example only and that modifications to detail may be made while still falling within the scope of the invention.
Example 1 - Construction of LAMP Constructs [0277] The improved LAMP Constructs illustrated in Figure 1 can be constructed using standard molecular biology techniques well known to the skilled artisan. For example, plasmids comprising the polynucleotides can be designed to generate the different structures ILC-1 to ILC-6 shown in Figure 1. The LAMP domains illustrated in Figure 1 can be derived from the amino acid sequences shown in Figures 3-10. Preferably the LAMP domains are derived from the human LAMP proteins shown in Figures 3-10. The boundaries of each domain can be derived from Figure 2A and Figure 2B. It is envisioned that the corresponding domains can also be cloned from the orthologous sequences by identifying the equivalent domains when compared to the human sequence. The cancer antigens can be cloned into the described LAMP Constructs either individually or in combination as described in Table 1.
Example 2 - Immune Response Evaluation of Mice to LAMP Constructs [0278] The ability of the improved LAMP Constructs as described in Example 1 can be tested for their ability to modulate an immune response. For example, Female BALB/c mice can be immunized i.d with 50ug of the improved LAMP Constructs and 5ug of GMCSF in lOOul PBS using nanopass on day 0, 14 and 28. Experiment will then be terminated 4 weeks after the last dose.[0279] Splenocytes (3xl05/well) are stimulated with antigenic protein (10ug/ml) in T cell media (RPMI with 10% heat inactivated FBS,1% penicillin/streptomycin, and IX 2-ME), supernatants are collected 72h after. Supernatants are diluted (400ul supernatant + 200ul T cell media) and cytokines are evaluated by ELISA. IL-10 or IL-4 production can be measured via ELISPOT assay.
Example 3 - Improved Antigen Presentation using LAMP Constructs [0280] Survivin is the smallest member of the Inhibitor of Apoptosis (IAP) family of proteins, involved in inhibition of apoptosis and regulation of cell cycle. These functional attributes make Survivin a unique protein exhibiting divergent functions i.e. regulating cell proliferation and cell death. Expression of Survivin in tumors correlates with not only inhibition of apoptosis and a decreased rate of cell death, but also resistance to chemotherapy and aggressiveness of tumors [1-6]. Therefore, Survivin is an important target for cancer vaccines and therapeutics [7-9]. Survivin has also been found to be prominently expressed on both human and embryonic stem cells and many somatic stem cell types indicating its yet unexplored role in stem cell generation and maintenance.
WO 2018/204534 PCT/US2018/030725 id="p-281" id="p-281" id="p-281"
[0281] Cancer is a heterogeneous group of diseases where abnormal cell growth with potential to invade other body parts takes control of normal homeostasis and becomes fatal if not timely and rightly treated. Immunotherapy specifically targets tumor cells thereby avoiding collateral damage to non-tumor cells and inducing anti-tumor response. This anti-tumor response also has the potential to eradicate tumor at distant sites in the body which may not be possible by surgical resection. Induction or enhancement of anti-tumor immune response is a formidable challenge in cancer because tumor cells use multiple evasion strategies and avoid being detected or eliminated by immune cells.[0282] The aim of this project is to evaluate in vivo immune response of all new generation of LAMP Constructs injected by I.D. in BALB/c mice. Specifically, mice were immunized with 50 pg of the tested constructs defined in the legend of Figure 1 by intradermal injection. No adjuvants were added at this experiment. Six mice per group were administrated with vaccines every 7 days with total three dose in one month. Immune response was monitored 14 days after the last immunization.[0283] The tested LAMP constructs were generated as described herein and the sequence of each tested construct is shown in Figure 19.Survivin protein was purchased from MyBiosource (San Diego, CA). Survivin peptides were from GenScript (Piscataway, NJ). Anti-survivin and m-IgGk-BP-HRP were bought from Santa Cruz Biotechnology (Dallas, TX), and mouse Monoclonal anti-LAMP-l/CD107a were from OriGene Technologies (Rockville, MD). ELIS POT antibody pairs for IFNy were from Biolegend. Fluorescently coupled CD3, CD4, CDS, CD44, CD62L, IFNy, TNFa, granzyme B, CD69 monoclonal antibodies and Zombie aqua fixable viability kit were purchased from BioLegend (San Diego, CA). Goat anti-mouse IgG2a-HRP and goat anti-mouse IgG-HRP were purchased from Southern Biotechnologies (Birmingham, AL). Streptavidin- HRP was purchased from Thermo Fisher (Waltham, MA). SureBlue TMB microwell peroxidase substrate and TMB stop solution were purchased from KPL (Gaithersburg, MD).[0284] 50 pg of each construct was used in a total volume of 100 ul per mouse per dose for Pharmajet.Mice were immunized with the vaccine by i.d. delivery on days 0, 7, and 14. Mice were bled on days 28 for serum collection. Serum was collected and stored in -30°C. Spleens were collected on day 28 at the termination of experiment and processed for ELISPOT and FACS to evaluate survivin specific T cell responses.[0285] Measurement of plasma survivin-specific total IgG by ELISA.The murine antibody response to survivin was assessed by indirect ELISA. ELISA plates (MaxiSorp) were coated with 2 ug/ml survivin (1-142) protein in carbonate-bicarbonate buffer overnight and then blocked with 2% BSA in PBS. Plasma samples were diluted 1:100 in blocking buffer. Samples were detected with goat anti-mouse IgG-HRP (Southern Biotech, Birmingham, AL). Reaction was developed with SureBlue TMB Substrate and stopped with TMB Stop Solution from KPL (Gaithersburg, MD). Plates were read (OD450) by using Epoch ELISA reader (BioTek, Winooski, VT).[0286] Evaluation of antigen-specific T cell response.To assess antigen-specific T cell response in the vaccinated mice, splenocytes from vaccinated mice were evaluated for antigen-specific IFNy production by WO 2018/204534 PCT/US2018/030725 Enzyme-linked immunospot (ELISPOT). For ELISPOT assays, 96-well nitrocellulose plates (Millipore), were coated overnight at 4°C with 100 pl/well of capture monoclonal antibody in PBS. The plates were washed three times with 200 pl/well PBS and blocked with 200 pl /well T cell media for at least 2 hrs at room temperature. Splenocytes were plated at 3xl05 cells/well and co-cultured with 2 ug/ml pooled peptides of Survivin (Table 2) or concavalin A (0.125 ug/ml) or medium alone in a total volume of 200 pl/well T cell media (RPMI-1640 with L-Glutamine and HEPES (ATCC), 1% penicillin, 1% streptomycin, and 5 x 10 5M B-ME) at 3xl05 cells/well for 48h at 37°C in 5% CO2- The plates were washed two times with 200 p l/wcll PBS and two times with 2pl/well PBS-T (0.05% Tween/PBS). Diluted detection antibodies (50 pl/well in PBS-T/0.5%BSA) were added and plates were incubated for 2 hrs with shaking at room temperature. Plates were washed four times with PBS. Streptavidin-alkaline phosphatase diluted in PBS (50 pl/well) were added and incubated for 2 h. Plates were washed with PBS four times and developed with 50 pl/well of 3-Amino-9-Ethylcarbazole (AEC, BD Bioscience) substrate for 10 min. Color development was stopped by washing under running tap water. After drying 72h at room temperature in dark, colored spots were counted using an AID ELISPOT High-Resolution Reader System and AID ELISPOT Software version 3.5 (Autoimmun Diagnostika GmbH).
Table 2. Pooled peptic es from GenscriptPooled Pl Surl-15, Suri 1-25, Sur 21-35, Sur31-45, sur 41-55Pooled P2 Sur51-65, sur61-75, sur71-85, sur81-95Pooled P3 Sur91-105, sur 101-115, surl 11-125, surl21-135, surl31-142Pooled P4 Sur31-45, sur41-55 and sur51-65 id="p-287" id="p-287" id="p-287"
[0287] Western blots.293T cells were transfected with the tested constructs using lipofectamine 2000 reagents (Invitrogen). Transfected cells were washed with PBS and suspended in 200 pl of RIPA lysis buffer with halt proteinase inhibitors (Thermo Scientific, Waltham, MA). Lysates were centrifuges (700 g for minutes at 4°C), followed by measurement of protein concentration in the clarified supernatants using Pierce BCA protein Assay kit (ThermoFisher Scientific, Waltham, MA). 10 pg of protein was electrophoresed in pre- cast (4-20%) SDS-PAGE gels (BioRad, Hercules, California), and transferred onto nitrocellulose membranes (BioRad) and immunoblotted with mAbs to hLAMP. Membranes were blocked with Detection™ block buffer (KPL) and probed with rabbit anti-human LAMP (Sino Biological Inc., Beijing, China) or anti-survivin antibody and goat anti-rabbit-HRP antibody, and then developed with TMB (KPL).[0288] Flow cytometry.Cells were first labelled with Zombie aqua fixable viability dye in PBS (1:500 dilution), followed by surface antibodies (1:100 dilution) in staining buffer (4% FBS, 2% rat serum, 2% mouse serum in PBS). For intracellular staining cells were stained with Zombie aqua, followed by surface staining, fixation with 4% paraformaldehyde, and stained with intracellular antibody in permeabilization buffer (PBS with 1% FCS 0.1% saponin). Samples were analyzed on a CytoFlex flow cytometer (Beckman Coulter) and analyzed using Kaluza software (Beckman Coulter).
WO 2018/204534 PCT/US2018/030725 id="p-289" id="p-289" id="p-289"
[0289] Statistics.Two-Way ANOVA test was performed using GraphPad Prism 6.0 software or R file to evaluate the statistical significance. Each mouse’s RPMI result was deducted from the results of the antigen activation.[0290] Vaccines, and immunizations.Control, Survivin+Complete LAMP, Survivin-ILC-Construct, Survivin-ILC-2, Survivin-ILC-3, and Survivin-ILC-4 Construct vaccines were used in a total volume of 25pl per mouse per dose. Mice were immunized with the vaccine by ID delivery on days 0, 7, and 14. Mice were bled on days -13, and 28 for serum collection. Serum was collected and stored in -30°C. Spleens were collected on day 28 and processed for ELISPOT/ELISA assays.[0291] Figure 14: Validation of the plasmids:293T cells were transfected with the plasmids for days. Transfected cells were lysed, and then electrophoresed in pre-cast SDS-PAGE gel. The proteins were transferred to nitrocellulose membranes and immunoblotted with mAbs to human LAMP (OriGene, #TA337108) or survivin Santa Cruz #17779). Molecular weight of LAMP = 100KD, Survivin = 16KD. Figure shows that all tested LAMP constructs produced appropriately sized protein.[0292] Figures 15 and 16: Tested LAMP Constructs induce Thl effector T cells producing IFNy. Female BALB/c mice were immunized i.d with 50 pg of the indicated constructs in 100 pl PBS via Pharmajet device on day 0, 7 and 14. Experiment was terminated 14 days after the last dose. Splenocytes (3xl05/well) were stimulated with survivin pooled peptides (4 ug/ml) in T cell media (RPMI with 10% heat inactivated FBS,1% penicillin/streptomycin, and IX -ME), for 48 h. A. IFNy production by spots. B. IFNy production induced by all pooled peptides (bar figure from A). n= 6 per group. Two way ANOVA (R file) was used for statistical analysis. Figure 14 shows that all tested LAMP constructs induced a robust T cell response as shown by IFNy production.[0281] We unexpectedly found that after 3 dose of the improved LAMP Constructs (one week apart), a robust Thl type response elicited by tested LAMP Constructs, especially ILC-4 where the hinge sequence was replaced by Survivin gene. More interestingly, improved LAMP Construct ILC-4 appears to recognize the Survivin epitopes from N-terminal to C-terminal, and induce T cell response against human Survivin peptide sequence which is 100% identical to the mouse. We also found longer (72hrs) stimulation of frozen-thawed splenocyte cells with Survivin peptides, ILC-4 showed significant higher IFNy production than the first generation of LAMP-Survivin (see Figure 19). Specifically, Figure 16shows that the all improved LAMP Constructs tested showed higher T cell response with ILC-4 having the best activity as this constructed elicited a significantly higher T cell response against all Survivin peptides pools. Moreover, contrary to what was known in the art, removal of the second homology domain of the luminal domain created an improved LAMP construct that elicited a more robust immune response as compared to the complete LAMP construct (see, results for ILC- and ILC-3). Frozen splenocytes (4xl05/well) were stimulated with pooled peptides 4 (4 ,ug/ml) in T cell media (RPMI with 10% heat inactivated FBS,1% penicillin/streptomycin, and IX -ME), for 48 h. n=6 per group. Two way ANOVA was used for statistical analysis. *p<0.05, **p<0.01, ***p<0.005, ****p<0.0001 WO 2018/204534 PCT/US2018/030725 id="p-293" id="p-293" id="p-293"
[0293] Figure 17. CD4 T cells are the major source of IFNy producing cells.Female B ALB/c mice were immunized i.d with 50 pg of the indicated vaccines in 100 pl PBS via Pharmajet device on day 0, 7 and 14. Experiment was terminated 14 days after the last dose. Splenocytes (lxl06/well) were stimulated with pooled peptides 1 (4 ug/ml) in T cell media (RPMI with 10% heat inactivated FBS,1% penicillin/streptomycin, and IX P־ME) over night, followed by adding monesin and brefeldin A and culturing for additional 5h. Cells were harvested and stained by Zombie, surface marker, and intracellular staining according to ITI staining protocol. Cells are gated on memory CD4 T cells (CD4+CD44+CD62L-) or CDS T cells (CD8+CD44+CD62L- ). Data is representative of one mouse in each group. While there is an increase in CDS effector memory cells in vaccinated mice with the various constructs, IFNy production is more pronounced in the CD4 T cell population.[0294] Figure 18: Improved LAMP Constructs produced stronger Survivin-specific total IgG response in BALB/c mice.Female B ALB/c mice were immunized i.d with 50 pg of the indicated vaccines in 100 pl PBS via Pharmajet device on day 0, 7 and 14. Experiment was terminated 14 days after the last dose. Mice were bleed on days 28. Serum was separated and stored in -30°C. Total IgG and IgG2a were determined in serum by ELISA. Briefly, ELISA plates were coated with 2 ug/ml Survivin (l-142aa), blocked with PBS/2%BSA, serum (1:100 dilution in blocking buffer) were evaluated by HRP-conjugated goat anti mouse IgG (1:6000) and IgG2a (1:11000). n=6 mice per group. **p<0.01, ***p<0.005, ****p<0.0001. Importantly and contrary to what was known in the art, Figure 18shows that fragments of the luminal domain worked better than use of the complete luminal domain (i.e., compare complete LAMP construct with constructs ILC-2 and IL-3). Unexpectedly, insertion of the antigen between the two homology domains of the luminal domain generated the strongest antibody response (see, ILC-4).[0295] References relied on in this section. 1. Kami K, Doi R, Koizumi M, Toyoda E, Mori T, Ito D, et al. Survivin expression is a prognostic marker in pancreatic cancer patients. Surgery. 2004;136(2):443-8. doi: 10.1016/j.surg.2004.05.023. PubMed PMID: 15300213.2. Zhang SQ, Qiang SY, Yang WB, Jiang JT, Ji ZZ. [Expression of survivin in different stages of carcinogenesis and progression of breast cancer]. Ai Zheng. 2004;23(6):697-700. PubMed PMID: 15191674. 3. Zhang X, Zhong L, Hu K, Li Q. [Expression of survivin and its correlation with apoptosis in non-small cell lung cancer]. Zhongguo Fei Ai Za Zhi. 2004;7(2):138-41. doi: 10.3779/j.issn.l009-3419.2004.02.14. PubMed PMID: 21215009.4. Kishi H, Igawa M, Kikuno N, Yoshino T, Urakami S, Shiina H. Expression of the survivin gene in prostate cancer: correlation with clinicopathological characteristics, proliferative activity and apoptosis. J Urol. 2004;171(5):1855-60. doi: 10.1097/0Lju.0000120317.88372.03. PubMed PMID: 15076293.
WO 2018/204534 PCT/US2018/030725 . Asanuma K, Tsuji N, Endoh T, Yagihashi A, Watanabe N. Survivin enhances Fas ligand expression via up-regulation of specificity protein !-mediated gene transcription in colon cancer cells. J Immunol. 2004;172(6):3922-9. PubMedPMID: 15004200.6. Miyachi K, Sasaki K, Onodera S, Taguchi T, Nagamachi M, Kaneko H, et al. Correlation between survivin mRNA expression and lymph node metastasis in gastric cancer. Gastric Cancer. 2003;6(4):217-24. doi: 10.1007/510120-003-0255-2. PubMedPMID: 14716515.7. Badana AK, Chintala M, Gavara MM, Naik S, Kumari S, Kappala VR, et al. Lipid rafts disruption induces apoptosis by attenuating expression of LRP6 and survivin in triple negative breast cancer. Biomed Pharmacother. 2017;97:359-68. doi: 10.1016/j.biopha.2017.10.045. PubMedPMID: 29091885.8. Cai IP, Wang YD, Zhang X, Xue HZ. [Expression of P16 and survivin in liver cancer and their clinical significance]. Zhonghua Gan Zang Bing Za Zhi. 2017;25(10):778-80. doi: 10.3760/cma.j.issn.l007- 3418.2017.10.013. PubMedPMID: 29108210.9. Cho HI, Kim HR, Park YS, Kim YH, Kim DK, Park SI. Prognostic value of survivin expression in stage III non-small cell lung cancer patients treated with platinum-based therapy. Surg Oncol. 2015 ;24(4) :329- 34. doi: 10.1016/j.suronc.2015.09.001. PubMedPMID: 26690822.10. Godinho RM, Matassoli FL, Lucas CG, Rigato PO, Goncalves JL, Sato MN, et al. Regulation of HIV- Gag expression and targeting to the endolysosomal/secretory pathway by the luminal domain of lysosomal- associated membrane protein (LAMP-1) enhance Gag-specific immune response. PLoS One. 2014;9(6):e99887. doi: 10.1371/journal.pone.0099887. PubMed PMID: 24932692; PubMed Central PMCID: PMCPMC4059647.
Example 4: Therapeutic Treatment of LAMP Constructs [0296] Female BALB/c mice can be inoculated s.c with syngeneic 7000 4T1 mammary carcinoma cells on day 0. Vaccine 50ug and 5ug of GMCSF in lOOul PBS is given i.d using nanopass once the tumors are palpable. Primary tumors are measured with a caliper and tumor volume is calculated using the formula p/(length x width)3/2. Average tumor volume as a function of days after tumor inoculation can be measured. A Kaplan-Meier plot can be used to show overall survival at the point of termination.
Example 5 - Prime/Boost Protocol [0297] Herpesvirus entry mediator (HVEM), also known as tumor necrosis factor receptor superfamily member 14 (TNFRSF14) or CD270, is a human cell surface receptor of the TNF-receptor superfamily. In recent years, HVEM has been found highly expressed on hematopoietic cells and a variety of parenchymal cells, such as breast, melanoma, colorectal, and ovarian cancer cells, as well as gut epithelium. HVEM is a bidirectional protein, either inhibiting or stimulating T cells, through binding to BTLA or LIGHT (TNFSF14).
WO 2018/204534 PCT/US2018/030725 id="p-298" id="p-298" id="p-298"
[0298] We generated a DNA vaccine encoding HVEM-LAMP to generate an antibody which could block the inhibitory function of HVEM for tumor therapeutic applications. We hypothesized that LAMP will promote the antibody response by enhancing the affinity of HVEM specific antibodies and/or expanding the repertoire of B cell epitopes in the HVEM protein. In this study, we compared the immunogenicity of HVEM encoding plasmid with and without LAMP (SEQ ID NO: 158 and SEQ ID NO: 159). Plasmids encoding HVEM-LAMP and HVEM and recombinant HVEM protein were designed as described herein.[0299] Goat anti-mouse IgG-HRP was purchased from Southern Biotechnologies (Birmingham, AL). SureBlue TMB microwell peroxidase substrate and TMB stop solution were purchased from KPL (Gaithersburg, MD). ELISPOT plates were ordered from EMD Millipore (Billerica, MA, Cat. No. MAIPS4510). IFN-y antibody pair used in ELISPOT was purchased from BioLegend (San Diego, CA) and clones AN 18 and R46A2 were used as coating and detection, respectively. Streptavidin-HRP and AEG substrate were purchased from BD Biosciences (San Jose, CA).[0300] Six to eight week old female Balb/c mice were purchased from Harlan Laboratories (Frederick, MA) and maintained at animal facility in Immunomic Therapeutics, Inc. (Rockville, MA). Mice (n=6) were treated with lOpg/dose of HVEM-LAMP, HVEM, or LAMP vector control by electroporation IM delivery at days 0, 7, and 14. On day 35, mice were boosted with 5pg HVEM protein in the presence of Alum by i.p. injection. On day 28 and 49, mice were bled and sera were isolated for antibody detection. Mice were sacrificed on day 56 and splenocytes were tested for IFN-y production by ELISPOT.[0301] ELISA procedure was followed by Su et al., J of Immunol Res; (10): 1 -15 (2016). Plates were coated with 5pg/ml HVEM protein. Data were analyzed by using Microsoft Excel and Prism 6 software.[0302] The primary aim of this study was to compare the antibody profiles between HVEM-LAMP and HVEM. On day 28, HVEM-LAMP vaccinated mice produced significant higher level of HVEM specific IgG antibody than that of the HVEM group (Figure 11).After a protein boost, the HVEM specific antibody was increased about 1000-fold in HVEM immunized mice and the mean titer was changed from 100 to 108000. This result indicates that the immune memory was induced by the HVEM DNA plasmid. Although HVEM DNA alone only induced a minimal antibody response, protein boost rapidly recalled the immune memory. On the other hand, HVEM-LAMP group again exhibited a significant higher titer than the HVEM and LAMP groups, the mean titer is 5 folds of the HVEM group, indicating the power of LAMP in enhancing antibody response (Figure 12). [0303] Additionally serum samples (Day 49) from HVEM+LAMP or HVEM alone immunized/HVEM protein boosted mice were pooled and tested for peptide mapping. Twelve peptides were found to be bound to the pooled serum (mouse IgG reaction) and seven of the twelve peptides showed strong binding affinity. HVEM+LAMP alters the binding affinity of peptides 17. 24,25, and 28 as compared to HVEM alone as shown in Figure 13.These changes may have physiological effects in protecting tumor growth.
WO 2018/204534 PCT/US2018/030725 id="p-304" id="p-304" id="p-304"
[0304] In conclusion, data from this study suggest that two constructs were expressed in vivo and LAMP significantly improved the humoral immune response.
Example 6: Production of an Antibody from a Polypeptide [0305] Anti-antigen antibodies can be prepared by a variety of standard methods of raising antibodies using animal injection. (See, Current Protocols, Chapter 2.) For example, cells expressing an improved LAMP Construct comprising a cancer antigen described herein is administered to a non-human vertebrate to induce the production of sera containing polyclonal antibodies. In a preferred method, a preparation of the LAMP/antigen protein is prepared and purified to render it substantially free of natural contaminants. Such a preparation is then introduced into the non-human vertebrate to produce polyclonal antisera of greater specific activity.[0306] In the most preferred method, the anti-antigen antibodies of the present invention are monoclonal antibodies (or protein binding fragments thereof). Such monoclonal antibodies can be prepared using hybridoma technology. (Kohler et al., Nature 256:495 (1975); Kohler et al., Eur. J. Immunol. 6:5(1976); Kohler et al., Eur. J. Immunol. 6:292 (1976); Hammerling et al., in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., pp. 563-681 (1981).) In general, such procedures involve immunizing a non- human vertebrate animal (preferably a rabbit, mouse, cow, camel, llama) with an improved LAMP Construct comprising an antigen, the encoded polypeptide of an improved LAMP Construct comprising an antigen or, more preferably, with an improved LAMP Construct-expressing cell. Such cells may be cultured in any suitable tissue culture medium; however, it is preferable to culture cells in Earle's modified Eagle's medium supplemented with 10% fetal bovine serum (inactivated at about 56 degrees C), and supplemented with about g/1 of nonessential amino acids, about 1,000 U/ml of penicillin, and about 100 ug/ml of streptomycin.[0307] The splenocytes of such non-human vertebrate host (e.g, mice) are extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP20), available from the ATCCTM. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al. (Gastroenterology 80:225-232 (1981).) The hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding the antigen.[0308] It will be appreciated that Fab and F(ab')2 and other fragments of the anti-antigen antibodies may be used according to the methods disclosed herein. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). Alternatively, secreted protein-binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry.
WO 2018/204534 PCT/US2018/030725 id="p-309" id="p-309" id="p-309"
[0309] For in vivo use of antibodies in humans, it may be preferable to use "humanized" chimeric monoclonal antibodies. Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric antibodies are known in the art. (See, for review, Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Cabilly et al., U.S. Pat. No. 4,816,567; Taniguchi et al., EP 171496; Morrison et al., EP 173494; Neuberger et al., WO 8601533; Robinson et al., WO 8702671; Boulianne et al., Nature 312:643 (1984); Neuberger et al., Nature 314:268 (1985).) Example 7: Use of Polynucleotides to Generate Polyclonal and Monoclonal Antibodies [0310] Methods of directly injecting polynucleotides into animals are well described in the art. See, for example, U.S. patent numbers 5,676,954; 6,875,748; 5,661,133. For example, a polynucleotide encoding an improved LAMP Construct comprising an antigen can be injected into the quadriceps muscles of restrained awake mice (female 6-12 week old BALB/c or Nude, nu/nu, from Harlan Sprague Dawley, Indianapolis, Ind.). In one embodiment, 50 pg of a polynucleotide in 50 pl solution using a disposable sterile, plastic insulin syringe and 28G 1/2 needle (Becton-Dickinson, Franklin Lakes, N.J., Cat. No. 329430) fitted with a plastic collar cut from a micropipette tip can be used to inject the mice, as described in Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996)).[0311] Alternatively, 6-week old Sprague Dawley female mice (body weight 20-25 grams) can be given 5000 ppm ZnOSO4 in their drinking water beginning 24 hours prior to injection. This amount of zinc has been shown to be able to activate the metallothionein promoter. Each mouse is then injected intravenously through a tail vein puncture with a 25 gauge needle with 30 pg of a polynucleotide encoding an improved LAMP Construct comprising an antigen complexed with 150 pg liposome (Lipofection TM) in a total volume of 30 pl. Animal care should be maintained throughout the study and should be performed in compliance with the "Guide for the Use and Care of Laboratory Animals", Institute of Laboratory Animal Resources, Commission on Life Sciences, National Research Council, National Academy Press.[0312] After the injected polynucleotide encoding the improved LAMP Construct comprising an antigen is delivered into the cells in the animal, the antigen is delivered to the endosome/lysosome, processed and presented to the immune system. The improved LAMP Construct comprising an antigen can then stimulate the production of antibodies specific to the antigen. These antibodies can be isolated and used as a polyclonal mixture or further isolated into single species or monoclonals. The process of the immune response and production of antibodies against foreign antigens in vivo are well known in the art.[0313] In a third animal model, Balb/c 3T3 A31 cells are transfected by electroporation with a polynucleotide encoding an improved LAMP Construct comprising an antigen. G418 resistant clones expressing LAMP Construct comprising an antigen are identified by their ability to bind human RBC. To generate polyclonal antibodies, Balb/c mice are immunized twice intraperitoneally, at an interval of 14 days, WO 2018/204534 PCT/US2018/030725 with 107 cells comprising the improved LAMP Construct comprising an antigen. After a final boost, the immune serum is collected, IgG is purified by protein G Sepharose and passed over an antigen column prepared by coupling 1.0 mg purified antigen to cyanogen bromide activated Sepharose CL-4B. Bound IgG can be eluted with 0.1 M glycine buffer pH 2.5 and neutralized with 0.1 volumes of 0.1 M Tris pH 8.0. To generate a monoclonal antibody (mAb), Balb/c mice are immunized with LAMP Construct comprising an antigen and hybridomas are generated by fusing immune spleen cells with the SP2 myeloma following standard methods (28). A positive well reacting specifically with an antigen can be identified by enzyme-linked immunosorbent assays as described in the art. The hybridoma is cloned three times by limiting dilution to produce an antibody.
Example 8: Immunization of an Improved LAMP Construct Comprising an Antigen [0314] Methods of raising antibodies in mammals are well known in the art. In one example, polyclonal antiserum against a LAMP Construct comprising an antigen is raised by immunization of pathogen free rabbits with a total of 500 pg of an improved LAMP Construct comprising an antigen over a period of two months. For example, the improved LAMP Construct comprising an antigen can be dissolved in PBS and emulsified with an equal volume of Freund’s adjuvant. After the final booster, the serum of the rabbits can be separated to determine the titer of the polyclonal antiserum.[0315] In an additional animal model, groups of 5 mice (C57BL/6J; Jackson Labs) can be subcutaneously immunized with 5 pg of endotoxin-free LAMP Construct comprising an antigen emulsified in alum. Three weeks later, mice are bled and the presence of anti-antigen specific antibodies can be determined by titering the seras by ELISA (direct binding of antibodies in sera to wild type BPTI or APP-KI coated, directly or indirectly (via a biotinylated tag and streptavidin), on the wells).[0316] To obtain monoclonal antibodies, 4-6 week old Balb/c mice can be immunized with an improved LAMP Construct comprising an antigen (for example 4 times with 2 week intervals with 10-1pg/injection dissolved in Freunds complete adjuvant for the first injection, and Freund’s incomplete adjuvant for subsequent immunizations). Splenocytes are isolated and fused with a fusion cell line such as Sp2/myeloma cells, followed by limiting dilution. Growing clones are screened using for example an enzyme- linked immunosorbant assay (ELISA). 96 cells plates are coated with an improved LAMP Construct comprising an antigen or with a control protein. The culture supernatant is added, followed by washing and addition of a labeled anti-mouse antibody for detection. After limited dilution cloning of the anti-antigen antibody producing stable hybridomas are obtained. From each cell, supernatant is collected and by affinity chromatography using protein A sepharose columns monoclonal antibodies can be purified.
Example 9 - LAMP Constructs with IGFBP-2 [0317] Cancer vaccine development has been pursued for more than 4 decades. However, despite numerous clinical studies of a variety of cancer vaccine formulations, therapeutic cancer vaccination has had WO 2018/204534 PCT/US2018/030725 overall clinical benefit of about 20%L 25 ׳ 4 ׳ 3 ׳ . Obstacles to improve efficacy have included a) poor immunogenicity of self-proteins that are cancer associated antigens, b) induction of regulatory T cells6, and c) low immune response obtained by vaccination. Recent reports suggest a correlation of vaccine-induced Thl immune infiltrate in tumor microenvironment with improved clinical response in many tumors7,811 ׳ 10 ׳ 9 ׳ .[0318] Insulin-like growth factor binding protein 2 (IGFBP-2) is a self-protein elevated during progression of many human tumors. IGFBP-2 overexpression is correlated with high-grade gliomas with poor prognosis12 and is an independent indicator of malignancy in breast cancer patients13. IGFBP2 is associated with metastasis and poor survival in lung cancer patients14. IGFBP2 overexpression is also correlated with advanced disease in adrenal cancer15, bladder cancer16, gastric cancer17, ovarian cancer18, 19, and prostrate cancer ’ MATERIALS AND METHODS [0319] Mice.Six to eight week old female C57BL/6 albino mice were purchased from Envigo and maintained at animal facility in Immunomic Therapeutics, Inc. (Rockville, MA).[0320] Reagents and antibodies.Control vector Control, IGFBP2 (l-328)-no LAMP, and IGFBP(39-328)-ILC-l Construct were made as described herein using standard protocols. Anti-mouse rabbit IGFBPPoAb was from MyBiosource (San Diego, CA), goat anti-rabbit IgG-FITC was from e-biosciences (San Diego, CA), rabbit anti-human LAMP was from Sino Biological (Beijing, China). Goat anti-rabbit-HRP, goat anti- mouse IgGl-HRP and goat anti-mouse IgG2a-HRP were purchased from Southern Biotechnologies (Birmingham, AL). Streptavidin-HRP were purchased from Thermo Fisher (Waltham, MA). SureBlue TMB microwell peroxidase substrate and TMB stop solution were purchased from KPL (Gaithersburg, MD). ELISPOT antibody pairs for IFNy, and IL-10 was from BioLegend. Cytokine ELISA kits were from R&D systems.[0321] Vaccines, and immunizations.Control, IGFBP2 (39-328)-ILC-l Construct, and IGFBP2 (1- 328)-no LAMP vaccines were used in a total volume of 20pl per mouse per dose. Mice were immunized with the vaccine by IM delivery via electroporation on days 0, 7, and 14. Mice were bled on days -13, and 28 for serum collection. Serum was collected and stored in -30°C. Spleens were collected on day 28 and processed for ELISPOT/ELISA assays.[0322] Measurement of serum IGFBP-2-specific IgG by ELISA.The murine antibody response to IGFBP-2 was assessed by indirect ELISA. ELISA plates (MaxiSorp) were coated with 5ug/ml IGFBP2 (39- 328) protein in carbonate bicarbonate buffer overnight and then blocked with 2% BSA in PBS. Serum samples were diluted (1:100, 1:300, 1:900, 1:2700, 1:8100, 1:24300, 1:72900, 1:218700) in PBS-T. Samples were detected with 1:6000 goat anti-mouse IgG-HRP (Southern Biotech, Birmingham, Al), followed by Streptavidin- HRP (Thermo Fisher Scientific, Rockford, IL). Reaction was developed with SureBlue TMB Substrate and stopped with TMB Stop Solution from KPL (Gaithersburg, MD). Plates were read (OD450) by using Epoch ELISA reader (BioTek, Winooski, VT). Average background (PBS only) was calculated, and samples which WO 2018/204534 PCT/US2018/030725 have OD450 value more than 2 * average background were considered as positive. The dilutions of such samples are determined as the endpoint titers.[0323] Evaluation of antigen-specific T cell response.To assess antigen-specific T cell response in the vaccinated mice, splenocytes from vaccinated mice were evaluated for antigen-specific IFNy, and IL-10 by Enzyme-linked immunospot (ELISPOT).[0324] Splenocytes were depleted of RBCs and co-cultured in flat bottom 96-well plates in 200pl/well T cell media (RPMI-1640 with L-Glutamine and HEPES (ATCC), 1% penicillin, 1% streptomycin, and 5 x 5M 2-ME) at 4xl05 cells/well and 10ug/ml IGFBP2 (39-328) protein or concavalin A (0.25pg/ml) or medium alone for 72h at 37°C in 5%CO2- Plates were centrifuged at 1600 rpm for 6 minutes and supernatants were collected and stored at -30°C.[0325] For ELISPOT assays were performed as described herein. Briefly, 96-well nitrocellulose plates(Millipore), were activated with 30ul of 35% ethanol for one minute, washed with PBS twice, and coated overnight at 4°C with SOpPwell of capture monoclonal antibody in PBS. The plates were washed three times with 200pl/well PBS and blocked with 200pl/well T cell media for at least 2hrs at room temperature. Splenocytes were plated at 4xl05 cells/well and co-cultured with 10ug/ml IGFBP2 (39-328) protein or 10ug/ml peptide pools or concavalin A (0.25pg/ml) or medium alone in a total volume of 200pl/well T cell media for 48h at 37°C in 5%CO2- The plates were washed, diluted detection antibodies (50pl/well) were added and plates were incubated for 2hrs at room temperature on shaker. Plates were washed four times with PBS. Streptavidin- HRP diluted in PBS (50pl/well) were added and incubated for Ih. Plates were washed with PBS and developed with 50pl/well AEG development solution for up to 30 min. Color development was stopped by washing under running tap water. After drying 24-72h at room temperature in dark, colored spots were counted using an AID ELISPOT High-Resolution Reader System and AID ELISPOT Software version 3.5 (Autoimmun Diagnostika GmbH).[0326] Evaluation of intracellular cytokine.To determine the cells responsible for production of cytokines intracellular staining was performed. Splenocytes were depleted of RBCs stored in refrigerator for 24h. Next day splenocytes were co-cultured in flat bottom 96-well plates in 200ul/well T cell media (RPMI- 1640 with L-Glutamine and HEPES (ATCC), 1% penicillin, 1% streptomycin, and 5 x 10-5M 2-ME) at 3xlcells/well and 20ug/ml IGFBP2 (39-328) protein or peptide pool or medium alone. A mixture of Brafeldin A and monensin (XI000) was added to each well in 20ul volume (0.22ul per well for 20ul volume; prepare a stock for all the wells) one hour later. PMA/ionomycin activation cocktail was added to positive control wells along with BEA and Monensin (Two wells were set-up; one for IgG control and other one for cytokine staining). Cells were incubated for total of 6h at 37°C in 5%CO2-[0327] Cells were washed with 200pl PBS. Cells were stained with 50ul of diluted Zombie aqua dye (1:500 dilution of zombie aqua in PBS) and incubated for 20 min at room temperature protected from light. Cells were washed once with 2%serum in PBS. 20ul of purified anti-mouse CD16/32mAb (clone 2.4G2; WO 2018/204534 PCT/US2018/030725 lug/20ul) was added to the cells and cells were incubated at 4°C for 10mins. Cells were centrifuged (2000 rpm, min), supernatant was decanted, and extracellular antibodies were added in PBS supplemented with 2% serum. The antibodies were 0.3 ul of each / well. Abs were added in a volume of 50pl/well, incubated for min in 4°C (the CD3 clone 17A2 can activate T cells so 4°C temperature is important).[0328] Cells were washed with PBS + protein (2% FCS), fixed and permeabilized with lOOpl of CytoFix/CytoPerm solution at 4°C for 30 mins (mixed thoroughly with the multichannel when adding the fix buffer to reduce aggregates and doublets formation). Cells were washed with 200pl Perm/Wash buffer (IX) twice (2000 rpm, 6 min). Cells were stained with the intracellular staining Ab at 0.5pl Ab/well in Perm buffer. One of the PMA/ionomycin wells were stained with the isotype controls. After Ih (or overnight) incubation cells were washed twice with perm buffer, and cells were acquired by flow cytometer. While acquisition the events/second were less than 8000. Acquired >106 cells per sample. The acquired data were analyzed using Kaluza analysis software. Dead cells and doublets were gated out.[0329] Statistics.Statistical analyses were performed by using Prism 6 software (GraphPad Software, La Jolla, CA). Data were analyzed by one-way ANOVA followed by Tukey’s test for multiple comparisons, p values below 0.05 were considered to indicate a statistically significant difference.[0330] Results. Figure 21:IGFBP2 (39-328)-ILC-l Construct induced significantly higher IFNy producing effector T cells at the dose of lOug IM/EP immunization. Peptide pool 1 induced the IFNy response similar to IGFBP-2 full length protein indicating dominant epitope in peptide pool 1. Splenocytes (4xl05/well) were stimulated with IGFBP-2 protein (10ug/ml) or peptide pool (10ug/ml) in T cell media (RPMI with 10% heat inactivated FBS,1% penicillin/streptomycin, and IX 2-ME), for 48h. Values in dot plot are experimental - media for each mouse. N=7 mice per group. Figure 21A.Dot plot to show recall with IGFBP2 full length (10ug/ml) and Figure 21B.Dot plot to show recall with peptide pool 1 (10ug/ml). * indicates p<0.05.[0331] Figure 22.IGFBP2 (39-328)-ILC-l Construct induced IFNy and/or TNFa producing CD4+ and CD8+ effector memory T cells at the dose of lOug IM/EP immunization. Splenocytes (4xl05/well) were stimulated with IGFBP-2 protein (10ug/ml) or peptide pool 1 (5ug/ml) in T cell media (RPMI with 10% heat inactivated FBS,1% penicillin/streptomycin, and IX 2-ME), for 6hr. Cells were stained for intracellular cytokines as described and analyzed as shown below. Representative data from each group.[0332] Figure 23: IGFBP-2-specific IgG production in C57BL/6 albino mice immunized with IGFBP2 (39-328)-ILC-l Construct by IM/EP.Female C57BL/6 albino mice were immunized IM/EP with lOug of the indicated vaccines in 20pl PBS on day 0, 7, and 14. Mice were bleed on days -6, 28. Serum was separated and stored in -30°C. IgG were determined in plasma by ELISA. Briefly, ELISA plates were coated with 5pg/ml IGFBP-2 (39-328) in carbonate bicarbonate buffer, blocked with 2%BSA, serum (1:100 dilution in PBS-T) were evaluated by HRP-conjugated goat anti mouse IgG (1:6000). Average background (PBS only) was calculated, and samples which have OD450 value more than 2 * average background are considered as positive. The dilutions of such samples are determined as the endpoint titers.
WO 2018/204534 PCT/US2018/030725 id="p-333" id="p-333" id="p-333"
[0333] Reference relied upon in this Example.1. Melero, I. et al. Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol 11,509-524 (2014). 2. Obeid, J., Hu, Y. & Slingluff, C.L., Jr. Vaccines, Adjuvants, and Dendritic Cell Activators—Current Status and Future Challenges. Semin Oncol 42,549-561 (2015). 3. Pol, J. et al. Trial Watch: Peptide-based anticancer vaccines. Oncoimmunology 4,0974411 (2015). 4. Speiser, D.E. & Romero, P. Molecularly defined vaccines for cancer immunotherapy, and protectiveT cell immunity. Semin Immunol 22,144-154 (2010).
. Thomas, A. & Giaccone, G. Why has active immunotherapy not worked in lung cancer? Ann Oncol 26,2213-2220 (2015). 6. Ebert, L.M. et al. A cancer vaccine induces expansion of NY-ESO-l-specific regulatory T cells in patients with advanced melanoma. PLoS One 7,048424 (2012). 7. Galon, J. et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313,1960-1964 (2006). 8. Fridman, W.H., Pages, F., Sautes-Fridman, C. & Galon, J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 12,298-306 (2012). 9. Renter, G.G. et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 361, 1838-1847 (2009).
. Madan, R.A., Bilusic, M., Heery, C., Schlom, J. & Gulley, J.L. Clinical evaluation of TRICOM vector therapeutic cancer vaccines. Semin Oncol 39,296-304 (2012). 11. Disis, M.L. et al. Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer. J Clin Oncol 27,4685-4692 (2009). 12. Moore, L.M. et al. IGFBP2 is a candidate biomarker for Ink4a-Arf status and a therapeutic target for high-grade gliomas. Proc Natl Acad Sci USA 106,16675-16679 (2009). 13. Busund, L.T. et al. Significant expression of IGFBP2 in breast cancer compared with benign lesions. J Clin Pathol 58,361-366 (2005). 14. Hu, Q. et al. Is insulin-like growth factor binding protein 2 associated with metastasis in lung cancer? Clin Exp Metastasis 31,535-541 (2014).
. Boulle, N., Logie, A., Gicquel, C., Perin, L. & Le Bouc, Y. Increased levels of insulin-like growth factor II (IGF-II) and IGF-binding protein-2 are associated with malignancy in sporadic adrenocortical tumors. J Clin Endocrinol Metab 83,1713-1720 (1998).
WO 2018/204534 PCT/US2018/030725 16. Miyake, H., Hara, I., Yamanaka, K., Muramaki, M. & Eto, H. Prognostic significance of insulin-like growth factor (IGF) binding protein-2 to IGF-binding protein-3 ratio in patients undergoing radical cystectomy for invasive transitional cell carcinoma of the bladder. BJU Int 95,987-991 (2005). 17. Shi, L.H., Zhu, X.Q., Zhao, G.H., Xia, Y.B. & Zhang, Y.S. Expression of insulin-like growth factor binding protein-2 in gastric carcinoma and its relationship with cell proliferation. World J Gastroenterol 12, 6285-6289 (2006). 18. Lee, E.J. et al. Insulin-like growth factor binding protein 2 promotes ovarian cancer cell invasion. Mol Cancer 4,7 (2005). 19. Lukanova, A. et al. Risk of ovarian cancer in relation to prediagnostic levels of C-peptide, insulin-like growth factor binding proteins-1 and -2 (USA, Sweden, Italy). Cancer Causes Control 14,285-292 (2003).
. Degraff, D.J., Aguiar, A.A. & Sikes, R.A. Disease evidence for IGFBP-2 as a key player in prostatecancer progression and development of osteosclerotic lesions. Am J Transl Res 1,115-130 (2009). 21. Shariat, S.F. et al. Association of preoperative plasma levels of insulin-like growth factor I and insulin-like growth factor binding proteins-2 and -3 with prostate cancer invasion, progression, and metastasis. J Clin Oncol 20,833-841 (2002).
Example 10: LAMP Constructs Comprising CMV pp65 and gB [0334] Glioblastoma multiforme (GBM) brain cancer is an aggressive human brain cancer with 5 year survival rates below 10% (Schuessler A et al. 2014). Current treatment includes surgical removal of the tumor and radiation, however successful treatment increases the median survival to only about 15 months. Several clinical trials are currently being evaluated for GBM, including immunotherapy. Interestingly, it was recently found that GBM tumors express CMV antigens including the antigen pp65. The role of CMV in GBM is unclear because CMV is not an oncogenic virus, however from a therapeutic perspective, the presence of CMV antigens provides a unique opportunity to exploit pre-existing antiviral immunity for immune-based GBM treatment. A recent report showed that autologous immunotherapy using DC transfected with LAMP-pp65 mRNA in a unique protocol significantly improved GBM patient’ survival. Specifically, Dr. Duane A. Mitchell from University of Florida, has shown that autologous DCs generated ex vivo, pulsed with LAMP-pp65 mRNA and injected back into GBM patients significantly improved the survival of GBM patients. The protocol for pp65- transfected DC also required pre-conditioning of the injection site by injection of Tetanus/Diphtheria vaccine 24h before DC injection (Mitchell DA et al, 2015).[0335] Here, we compare pp65+ILC-l LAMP construct with gB+ ILC-1 LAMP vaccine using PharmaJet and ID/EP.[0336] MATERIALS AND METHODS [0337] Reagents.Plasmid constructs Control, pp65+ILC-l LAMP construct, and gB+ ILC-1 LAMP were constructed as described herein. pp65 and gB peptides for ELISPOT were purchased from JPT (70% 91 WO 2018/204534 PCT/US2018/030725 purity). RBC lysis buffer was purchased from TonboBio and filtered through 0.22pm filter. ConA (Sigma) was used at 2.5ug/ml. Antibodies for IFNy ELISPOT were purchased from eBioscience or Biolegend. IL-ELISPTO reagents, SA-ALP and BCIP/NBT plus were purchased from MabTech. Ab Titers were evaluated using HRP anti- mouse antibodies from Southern Biotech. Lysate of cells infected by CMV was purchased from Virusys. The lysate was confirmed by the manufacturing company to be non-infective.[0338] Immunization and Plasma Collection.Six to eight week old male BALB/C mice bred and maintained at animal facility in Immunomic Therapeutics, Inc. Mice were immunized with the vaccine by either ID (Pharmajet) or ID via electroporation on days 0, 7, and 14. Mice were sacrificed on Day 26.[0339] ELISPOT.ELISPOT was carried out as described herein, with the exception of cell number: the cells were seeded at 2*105 splenocytes per well. Spleens from sacrificed mice were flushed with medium, red blood cells were lysed and then the cells were counted using a cellometer. 2*105 splenocytes were seeded per well of ELISPOT and incubated with medium (RPMI with HEPES + 10% HI equal Fetal +NaP + BME + P/S), duplicate of PP65 and gB peptides (1 ug/ml of each peptide) or ConA (2.5 ug/ml). Results are shown after averaging the duplicate and reducing the value from RPMI well for each mouse.[0340] Antibody titers.Antibodies titers were evaluated using a lysate of cells infected by CMV. Total IgG and IgG2a were evaluated. The results were only calculated for total IgG and only the 1:100 OD result was compared.[0341][0342] Results. Figure 24shows that IFNy was induced with both pp65 and gB peptides. Additionally, serum Ab were tested from the last bleed. When lysate from CMV-transfected cells was used, similar IgG titers are seen for both immunizations. Specifically, Figure 25shows that total IgG (left) and IgG2a (right) antibodies titer at in serum of immunized mice against CMV-transfected cell lysate. Statistical analysis was done in GraphPad prism, using Mann Whitney test. IgG2a CMV-lysate Ag - P = 0.041 Example 11. LAMP Constructs comprising PSMA [0343] Prostate cancer is the most frequently diagnosed cancer in men, and the second most common cause of cancer death in men in the US1. For localized disease, surgery and radiation continue to be curative treatments but have adverse effects such as urinary symptoms and sexual dysfunction that can negatively affect quality of life. For metastatic disease, chemotherapy as initial treatment now appears to extend survival compared with androgen deprivation therapy alone (ADT)2. ADT is associated with a) decreased bone mineral density and an increased risk of osteoporotic fragility fractures, b) development of castration resistant prostate cancer (CRPC). Methods to harness the immune system to recognize and kill prostate cancer cells are being exploited. One such attempt has led to Sipuleucel-T, a FDA approved, autologous cellular immunotherapy that targets prostatic acid phosphatase (PAP) for the treatment of asymptomatic or minimally symptomatic WO 2018/204534 PCT/US2018/030725 metastatic CRPC (mCRPC)3. Additional immunotherapies for prostate cancer are under development including a number of vaccine candidates, as well as approaches using targeted monoclonal antibodies4, 5.[0344] Several tumor-associated antigens (TAAs) have been identified that represent promising targets for T cell- or antibody-based immunotherapy. The group of molecules preferentially expressed in normal and malignant prostate tissues comprises prostate-specific membrane antigen (PSMA), prostein, prostate- specific antigen (PSA), prostatic acid phosphatase (PAP), prostate stem cell antigen (PSCA), T cell receptor gamma alternate reading frame protein (TARP), transient receptor potential (trp)-pS and six-transmembrane epithelial antigen of the prostate 1 (STEAP1).[0345] Human Prostate Specific Membrane Antigen (PSMA), also known as folate hydrolase I (FOLH1), is a 750-amino acid type II membrane glycoprotein, which is primarily expressed in normal human prostate epithelium and is upregulated in prostate cancer, including metastatic disease. Mouse homolog of PSMA (Folhl) is not expressed in the mouse prostate, but primarily expressed in the brain and kidney. Mouse Folhl encodes for a 752-amino acid protein, with 86% identity and 91% similarity to the human PSMA amino acid sequence6. PSMA is considered an important clinical biomarker of prostate because it is expressed many fold higher on prostate cells than cells of other tissues7,8,9. A strong correlation between increased PSMA expression and prostate cancer progression has been indicated10. Elevated PSMA expression is also associated with other malignancy including kidney and bladder cancer11. Elevated PSMA expression has been observed on tumor neovasculature, but not normal vasculature suggesting a role for PSMA in angiogenesis12. PMSA being a membrane protein is an attractive target to develop antibodies against it for diagnostic and therapeutic purposes13. Several therapeutic anti-PSMA mAbs have been developed, and many of these have been used in radioimmunotherapy for targeting cytotoxic radionucleotides, specifically to PSMA expressing cells14. Some anti-PSMA mAbs have been demonstrated to mediate a therapeutic effect by promoting an antibody-dependent cellular cytotoxicity (ADCC) effect that kills prostate cancer cells4,14,15[0346] MATERIALS AND METHODS [0347] Mice.Six to eight week old female C57BL/6 albino mice were purchased from Envigo and maintained at animal facility in Immunomic Therapeutics, Inc. (Rockville, MA).[0348] Reagents and antibodies.Control and PSMA-ILC-1 LAMP Constructs were made as described herein. Human PSMA PepMix (185 peptides, 15mers with llaa overlap) wasfromJPT (Acton, MA). Mouse anti-human PSMA Alexa Flour 488, Goat (Fab)2 anti-mouse IgG-Fc PE were from abeam (Cambridge, MA), rabbit anti-human LAMP was from Sino Biological (Beijing, China). Goat anti-rabbit-HRP, goat anti- mouse IgGl-HRP and goat anti-mouse IgG2a-HRP were purchased from Southern Biotechnologies (Birmingham, AL). Streptavidin-HRP were purchased from Thermo Fisher (Waltham, MA). SureBlue TMB microwell peroxidase substrate and TMB stop solution were purchased from KPL (Gaithersburg, MD). ELISPOT antibody pairs for IFNg was from BioLegend.
WO 2018/204534 PCT/US2018/030725 id="p-349" id="p-349" id="p-349"
[0349] Vaccines, and immunizations.Control vector, PSMA-ILC-1 LAMP Construct was used in a total volume of 20pl per mouse per dose. Mice were immunized with the vaccine by ID via electroporation on days 0, and 21. Serum was collected and stored in -30°C. Spleens were collected on day 34 for ELISPOT/ELISA assays.[0350] Evaluation of antigen-specific T cell response.To assess antigen-specific T cell response in the vaccinated mice, splenocytes from vaccinated mice were evaluated for antigen-specific IFNy and IL-10 by Enzyme-linked immunospot (ELISPOT). Splenocytes were depleted of RBCs and co-cultured in flat bottom 96-well plates in 200pl/well T cell media (RPMI-1640 with L-Glutamine and HEPES (ATCC), 1% penicillin, 1% streptomycin, and 5 x 10-5M 2-ME) at 4xl05 cells/well and 10ug/ml IGFBP2 (39-328) protein or concavalin A (0.25pg/ml) or medium alone for 72h at 37°C in 5%CO2- Plates were centrifuged at 1600 rpm for minutes and supernatants were collected and stored at -30°C.[0351 ] For ELISPOT assays were performed as described herein. Briefly, 96-well nitrocellulose plates(Millipore), were activated with 30ul of 35% ethanol for one minute, washed with PBS twice, and coated overnight at 4°C with SOpPwell of capture monoclonal antibody in PBS. The plates were washed three times with 200pl/well PBS and blocked with 200pl/well T cell media for at least 2hrs at room temperature. Splenocytes were plated at 3xl05 cells/well and co-cultured with 10, 5, 2, 1, 0.1, O.0lug/ml PSMA JPT peptide mix for PSMA or concavalin A (0.25pg/ml) or medium alone in a total volume of 200,111/we 11 T cell media for 48h at 37°C in 5%CO2- The plates were washed, diluted detection antibodies (50pl/well) were added and plates were incubated for 2hrs at room temperature on shaker. Plates were washed four times with PBS. Streptavidin- HRP diluted in PBS (50ul/well) were added and incubated for Ih. Plates were washed with PBS and developed with 50pl/well AEC development solution for up to 30 min. Color development was stopped by washing under running tap water. After drying 24-72h at room temperature in dark, colored spots were counted using an AID ELISPOT High-Resolution Reader System and AID ELISPOT Software version 3.5 (Autoimmun Diagnostika GmbH).[0352] Measurement of serum PSMA-specific IgG/IgG2a and Prostein-specific IgG/IgG2a by ELISA.The murine antibody response to PSMA was assessed by indirect ELISA. ELISA plates (MaxiSorp) were coated with 5ug/ml of PSMA protein in carbonate bicarbonate buffer overnight and then blocked with 2% BSA in PBS. Serum samples were diluted (1:100, 1:300, 1:900, 1:2700, 1:8100, 1:24300, 1:72900, 1:218700) in PBS-T. Samples were detected with 1:6000 goat anti-mouse IgG-HRP or 1:11000 goat anti mouse IgG2a- HRP (Southern Biotech, Birmingham, Al), followed by Streptavidin-HRP (Thermo Fisher Scientific, Rockford, IL). Reaction was developed with SureBlue TMB Substrate and stopped with TMB Stop Solution from KPL (Gaithersburg, MD). Plates were read (OD450) by using Epoch ELISA reader (BioTek, Winooski, VT). Average background (PBS only) was calculated, and samples which have OD450 value more than 2 * average background were considered as positive. The dilutions of such samples are determined as the endpoint titers.
WO 2018/204534 PCT/US2018/030725 id="p-353" id="p-353" id="p-353"
[0353] Evaluation of intracellular cytokine.To determine the cells responsible for production of cytokines intracellular staining was performed. Splenocytes were depleted of RBCs stored in refrigerator for 24h. Next day splenocytes were co-cultured in flat bottom 96-well plates in 200ul/well T cell media (RPMI- 1640 with L-Glutamine and HEPES (ATCC), 1% penicillin, 1% streptomycin, and 5 x 10-5M 2-ME) at 3xlcells/well and 2ug/ml PSMA (JPT peptide mix) or medium alone. A mixture of Brafeldin A and monensin (X1000) was added to each well in 20ul volume (0.22ul per well for 20ul volume; prepare a stock for all the wells) one hour later. PMA/ionomycin activation cocktail was added to positive control wells along with BEA and Monensin (Two wells were set-up; one for IgG control and other one for cytokine staining). Cells were incubated for total of 6h at 37°C in 5%CO2-[0354] Live/Dead staining (no protein in this step): Cells were washed with 200pl PBS (no protein to this wash). Cells were stained with 50ul of diluted Zombie aqua dye (1:500 dilution of zombie aqua in PBS) and incubated for 20 min at room temperature protected from light (no protein in this step). Cells were washed once with 2%serum in PBS.[0355] Pc block: 20ul of purified anti-mouse CD16/32mAb (clone 2.4G2; lug/20ul) was added to the cells and cells were incubated at 4°C for 10mins.[0356] Surface staining: cells were centrifuged (2000 rpm, 6 min), supernatant was decanted, and extracellular antibodies were added in PBS supplemented with 2% serum. The antibodies were 0.3 ul of each / well. Abs were added in a volume of 50pl/well, incubated for 30 min in 4°C (the CD3 clone 17A2 can activate T cells so 4°C temperature is important).[0357] Fixation and permeabilization: Cells were washed with PBS + protein (2% FCS), fixed and permeabilized with lOOpl of CytoFix/CytoPerm solution at 4°C for 30 mins (mixed thoroughly with the multichannel when adding the fix buffer to reduce aggregates and doublets formation). Cells were washed with 200pl Perm/Wash buffer (IX) twice (2000 rpm, 6 min)[0358] Intracellular staining: Cells were stained with the intracellular staining Ab at 0.5pl Ab/well in Perm buffer. One of the PMA/ionomycin wells were stained with the isotype controls. After Ih (or overnight) incubation cells were washed twice with perm buffer, and cells were acquired by flow cytometer. While acquisition the events/second were less than 8000. Acquired >106 cells per sample.[0359] The acquired data were analyzed using Kaluza analysis software. Dead cells and doublets were gated out.[0360] Statistics.Statistical analyses were performed by using Prism 6 software (GraphPad Software, La Jolla, CA). Data were analyzed by one-way ANOVA followed by Tukey’s test for multiple comparisons, p values below 0.05 were considered to indicate a statistically significant difference.[0361] Figure 26shows that the PSMA-ILC-1 LAMP Construct induced significantly higher IFNy producing effector T cells at the dose of 20ug ID/EP immunization. Splenocytes (3xl05/well) were stimulated with PSMA peptide mix from JPT (2, 1,0.1, 0.01, 0.001ug/ml) in T cell media (RPMI with 10% heat inactivated

Claims (28)

1./ Claims 1. A dendritic cell comprising a polynucleotide encoding a lysosomal associated membrane protein (LAMP) construct, wherein the LAMP Construct comprises two homology domains of a luminal domain of a LAMP protein, and an antigenic domain, wherein the antigenic domain is placed between the two homology domains, and wherein the antigenic domain comprises at least one epitope of pp65, at least one epitope of gB, and at least one epitope of IE1.
2. The dendritic cell of claim 1, wherein the LAMP Construct further comprises a transmembrane domain of a LAMP protein.
3. The dendritic cell of claim 1 or 2, wherein the LAMP Construct further comprises a signal sequence.
4. The dendritic cell of claim 3, wherein the signal sequence is derived from a LAMP protein.
5. The dendritic cell of any one of claims 1-4, wherein the LAMP Construct further comprises a cytoplasmic domain of a LAMP protein.
6. The dendritic cell of any one of claims 1-5, wherein the LAMP Construct comprises the structure of ILC-4.
7. The dendritic cell of any one of claims 1-6, wherein the antigenic domain is separated from one or both of the homology domains by a linker.
8. The dendritic cell of claim 7, wherein the linker is selected from the amino acid sequence GPGPG or PMGLP.
9. The dendritic cell of any one of claims 1-8, wherein the LAMP protein is selected from human LAMP-1, LAMP2, LAMP-3, LIMP 2, Macrosailin, Endolyn, LAMP5 or LIMBIC. 297073/
10. The dendritic cell of any one of claims 1-8, wherein the LAMP protein comprises an amino acid sequence selected from any one of SEQ ID NO:1-113.
11. The dendritic cell of any one of claims 1-8, wherein the LAMP protein comprises an amino acid sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical at least one of SEQ ID NO:1-113.
12. The dendritic cell of any one of claims 1-8, wherein the LAMP protein is human LAMP-1.
13. The dendritic cell of any one of claims 1-12, wherein the two homology domains comprise human LAMP-1 Homology Domain 1 and human LAMP-1 Homology Domain 2.
14. The dendritic cell of claim 13, wherein the human LAMP-1 Homology Domain comprises the amino acid sequence of residues 29-194 of SEQ ID NO: 1.
15. The dendritic cell of claim 13 or 14, wherein the human LAMP-1 Homology Domain comprises the amino acid sequence of residues 228-381 of SEQ ID NO: 1.
16. The dendritic cell of any one of claims 12-15, wherein the LAMP Construct further comprises a transmembrane domain comprising residues 383-405 of SEQ ID NO: 1.
17. The dendritic cell of any one of claims 12-16, wherein the LAMP Construct further comprises a cytoplasmic tail comprising residues 406-417 of SEQ ID NO: 1.
18. The dendritic cell of any one of claims 1-17, wherein the at least one epitope of ppcomprises the amino acid sequence of SEQ ID NO: 114, or comprises an amino acid sequence at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 114; and/or wherein the at least one epitope of gB comprises the amino acid sequence of SEQ ID NO: 117, or comprises an amino acid sequence at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 117; and/or wherein the at least one epitope of IE1 comprises the amino acid sequence of SEQ ID NO: 121, or comprises an amino acid 297073/ sequence at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 121.
19. The dendritic cell of any one of claims 1-17, wherein the at least one epitope of ppcomprises one or more of the amino acid sequences: LLQTGIHVRVSQPSL, ALPLKMLNIPSINVH, DQYVKVYLESFCEDV, IIKPGKISHIMLDVAFTSH, PQYSEHPTFTSQYRIQGKL, PPWQAGILARNLVPMV, or KYQEFFWDANDIYRIFA; and/or wherein the at least one epitope of gB comprises one or more of the amino acid sequences: TTSAQTRSVYSQHVT, QLIPDDYSNTHSTRYV, VSVFETSGGLVVFWQ, or NSAYEYVDYLFKRMIDLS; and/or wherein the at least one epitope of IE1 comprises one or more of the amino acid sequences: VLAELVKQIKVRVDMVRHRIKEHMLKKYTQ, IVPEDKREMWMACIKELH, KDELRRKMMYMCYRNIEFFTKNSAFPKTT, SVMKRRIEEICMKVFAQYI, AIAEESDEEEAIVAY, or VKSEPVSEIEEVAPEEEEDG.
20. The dendritic cell of any one of claims 1-17, wherein the antigenic domain comprises one or more of SEQ ID NO: 115, 116, 119, 120, 122, or 123.
21. The dendritic cell of any one of claims 1-17, wherein the antigenic domain comprises each of SEQ ID NO: 114, SEQ ID NO: 117, and SEQ ID NO: 121.
22. A composition comprising the dendritic cell of any one of claims 1-21.
23. The composition of claim 22 for use in reducing or treating diseases or disorders, in a subject in need thereof.
24. The composition of claim 22 for use in treating glioblastoma multiforme, in a subject in need thereof.
25. The composition of claim 22 for use in generating an immune response, in a subject in need thereof.
26. The composition for use of any one of claims 23-25, wherein the use comprises administering the composition to the subject in a priming step and/or at least one boosting step. 297073/ 1
27. The composition for use of claim 26, wherein the composition is administered in the priming step.
28. The composition for use of claim 26 or 27, wherein the composition is administered in at least one boosting step. For the Applicants REINHOLD COHN AND PARTNERS By:
IL297073A 2017-05-02 2018-05-02 Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens IL297073B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762500053P 2017-05-02 2017-05-02
US201762561760P 2017-09-22 2017-09-22
PCT/US2018/030725 WO2018204534A1 (en) 2017-05-02 2018-05-02 Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens

Publications (3)

Publication Number Publication Date
IL297073A IL297073A (en) 2022-12-01
IL297073B1 IL297073B1 (en) 2023-08-01
IL297073B2 true IL297073B2 (en) 2023-12-01

Family

ID=62528827

Family Applications (2)

Application Number Title Priority Date Filing Date
IL270271A IL270271B2 (en) 2017-05-02 2018-05-02 Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens
IL297073A IL297073B2 (en) 2017-05-02 2018-05-02 Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens

Family Applications Before (1)

Application Number Title Priority Date Filing Date
IL270271A IL270271B2 (en) 2017-05-02 2018-05-02 Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens

Country Status (10)

Country Link
US (1) US20200087365A1 (en)
EP (1) EP3618854A1 (en)
JP (3) JP7222915B2 (en)
KR (2) KR20240027895A (en)
CN (1) CN110913892A (en)
AU (1) AU2018263923A1 (en)
BR (1) BR112019023014A2 (en)
CA (1) CA3061950A1 (en)
IL (2) IL270271B2 (en)
WO (1) WO2018204534A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3793595A1 (en) * 2018-05-15 2021-03-24 Immunomic Therapeutics, Inc. Improved lamp constructs comprising allergens
WO2021077051A1 (en) * 2019-10-18 2021-04-22 Immunomic Therapeutics, Inc Improved lamp constructs comprising cancer antigens
US20220339245A1 (en) * 2021-04-27 2022-10-27 Academia Sinica Methods of treating hypertriglyceridemia or hypertriglyceridemia-related diseases
WO2023201201A1 (en) 2022-04-10 2023-10-19 Immunomic Therapeutics, Inc. Bicistronic lamp constructs comprising immune response enhancing genes and methods of use thereof

Family Cites Families (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4237115A (en) 1977-11-23 1980-12-02 Bactex, Inc. Method of immunization against enterotoxogenic infection by Escherichia coli
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4397844A (en) 1978-02-24 1983-08-09 Ciba-Geigy Corporation Antigen derivatives and processes for their preparation
GB2026340B (en) 1978-07-03 1982-12-22 Ash P Stabilising microvesicles
US4681762A (en) 1979-05-23 1987-07-21 Research Corporation Genetically attenuated bacterial vaccines with multiple mutations of the same phenotype
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
US4714681A (en) 1981-07-01 1987-12-22 The Board Of Reagents, The University Of Texas System Cancer Center Quadroma cells and trioma cells and methods for the production of same
US4769330A (en) 1981-12-24 1988-09-06 Health Research, Incorporated Modified vaccinia virus and methods for making and using the same
US4603112A (en) 1981-12-24 1986-07-29 Health Research, Incorporated Modified vaccinia virus
US4593002A (en) 1982-01-11 1986-06-03 Salk Institute Biotechnology/Industrial Associates, Inc. Viruses with recombinant surface proteins
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4592002A (en) 1983-12-13 1986-05-27 Honeywell Inc. Method of digital temperature compensation and a digital data handling system utilizing the same
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
ZA858044B (en) 1984-11-01 1987-05-27 American Home Prod Oral vaccines
WO1986005807A1 (en) 1985-04-01 1986-10-09 Celltech Limited Transformed myeloma cell-line and a process for the expression of a gene coding for a eukaryotic polypeptide employing same
FR2583429B1 (en) 1985-06-18 1989-11-03 Transgene Sa INTERFERON U EXPRESSION VECTOR IN MAMMALIAN CELLS, PROCESS FOR IMPLEMENTING SAME AND PRODUCT OBTAINED, AND PHARMACEUTICAL COMPOSITION CONTAINING INTERFERON U
AU606320B2 (en) 1985-11-01 1991-02-07 International Genetic Engineering, Inc. Modular assembly of antibody genes, antibodies prepared thereby and use
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US4737323A (en) 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
DE3642912A1 (en) 1986-12-16 1988-06-30 Leybold Ag MEASURING DEVICE FOR PARAMAGNETIC MEASURING DEVICES WITH A MEASURING CHAMBER
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
US4925648A (en) 1988-07-29 1990-05-15 Immunomedics, Inc. Detection and treatment of infectious and inflammatory lesions
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
US5744166A (en) 1989-02-25 1998-04-28 Danbiosyst Uk Limited Drug delivery compositions
EP0394827A1 (en) 1989-04-26 1990-10-31 F. Hoffmann-La Roche Ag Chimaeric CD4-immunoglobulin polypeptides
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
DE69029036T2 (en) 1989-06-29 1997-05-22 Medarex Inc SPECIFIC REAGENTS FOR AIDS THERAPY
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
US5676954A (en) 1989-11-03 1997-10-14 Vanderbilt University Method of in vivo delivery of functioning foreign genes
US5100587A (en) 1989-11-13 1992-03-31 The United States Of America As Represented By The Department Of Energy Solid-state radioluminescent zeolite-containing composition and light sources
DE69120146T2 (en) 1990-01-12 1996-12-12 Cell Genesys Inc GENERATION OF XENOGENIC ANTIBODIES
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
ES2129029T5 (en) 1990-10-05 2005-10-16 Celldex Therapeutics, Inc. DIRECT IMMUNOSTIMULATION WITH BISPECIFIC REAGENTS.
AU8727291A (en) 1990-10-29 1992-06-11 Cetus Oncology Corporation Bispecific antibodies, method of production, and uses thereof
US5179993A (en) 1991-03-26 1993-01-19 Hughes Aircraft Company Method of fabricating anisometric metal needles and birefringent suspension thereof in dielectric fluid
DE69214709T2 (en) 1991-04-26 1997-02-20 Surface Active Ltd New antibodies and methods of using them
SE503225C2 (en) 1991-10-30 1996-04-22 Leif Lindholm Konsult Ab Virus-antibody complex for introduction of virus into mammalian cells
US5661133B1 (en) 1991-11-12 1999-06-01 Univ Michigan Collateral blood vessel formation in cardiac muscle by injecting a dna sequence encoding an angiogenic protein
DK1024191T3 (en) 1991-12-02 2008-12-08 Medical Res Council Preparation of autoantibodies displayed on phage surfaces from antibody segment libraries
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
EP0627940B1 (en) 1992-03-05 2003-05-07 Board of Regents, The University of Texas System Use of immunoconjugates for the diagnosis and/or therapy of vascularized tumors
DE69325777T2 (en) 1992-06-09 2000-03-09 Hoppe Ag LATCH AND DOOR LOCK
GB9213033D0 (en) 1992-06-19 1992-08-05 British Telecomm Method of controlling a telecommunications network
US5747323A (en) 1992-12-31 1998-05-05 Institut National De La Sante Et De La Recherche Medicale (Inserm) Retroviral vectors comprising a VL30-derived psi region
US5633234A (en) 1993-01-22 1997-05-27 The Johns Hopkins University Lysosomal targeting of immunogens
FR2705686B1 (en) 1993-05-28 1995-08-18 Transgene Sa New defective adenoviruses and corresponding complementation lines.
FR2707091B1 (en) 1993-06-30 1997-04-04 Cohen Haguenauer Odile Retroviral vector for gene transfer and expression in eukaryotic cells.
US5928944A (en) 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
ZA951877B (en) 1994-03-07 1996-09-09 Dow Chemical Co Bioactive and/or targeted dendrimer conjugates
WO1995028494A1 (en) 1994-04-15 1995-10-26 Targeted Genetics Corporation Gene delivery fusion proteins
FR2719316B1 (en) 1994-04-28 1996-05-31 Idm New nucleic acid and polymer complexes, their preparation process and their use for cell transfection.
WO1996000295A1 (en) 1994-06-27 1996-01-04 The Johns Hopkins University Targeted gene delivery system
FR2722506B1 (en) 1994-07-13 1996-08-14 Rhone Poulenc Rorer Sa COMPOSITION CONTAINING NUCLEIC ACIDS, PREPARATION AND USES
FR2727689A1 (en) 1994-12-01 1996-06-07 Transgene Sa NEW PROCESS FOR THE PREPARATION OF A VIRAL VECTOR
US6030613A (en) 1995-01-17 2000-02-29 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
EP1323346B1 (en) 1995-01-17 2006-06-28 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of immunogens
US5837520A (en) 1995-03-07 1998-11-17 Canji, Inc. Method of purification of viral vectors
ES2304786T3 (en) 1995-04-27 2008-10-16 Amgen Fremont Inc. ANTI-IL-8 HUMAN ANTIBODIES, DERIVED FROM IMMUNIZED XENORATONES.
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
FR2737222B1 (en) 1995-07-24 1997-10-17 Transgene Sa NEW VIRAL AND LINEAR VECTORS FOR GENE THERAPY
WO1997035996A1 (en) 1996-03-25 1997-10-02 Transgene S.A. Packaging cell line based on human 293 cells
AU3447097A (en) 1996-07-01 1998-01-21 Rhone-Poulenc Rorer S.A. Method for producing recombinant adenovirus
JP2000514440A (en) 1996-07-09 2000-10-31 ザ ジョーンズ ホプキンス ユニバーシティー Gene transfer system
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
DK1500329T3 (en) 1996-12-03 2012-07-09 Amgen Fremont Inc Human antibodies that specifically bind TNF-alpha
JP2001506126A (en) 1996-12-13 2001-05-15 シェーリング コーポレイション Virus purification method
FR2759382A1 (en) 1997-02-10 1998-08-14 Transgene Sa NOVEL COMPOUNDS AND COMPOSITIONS CONTAINING THEM FOR USE IN TRANSFERRING AT LEAST ONE THERAPEUTICALLY ACTIVE SUBSTANCE, IN PARTICULAR A POLYNUCLEOTIDE, INTO A TARGET CELL AND USE IN GENE THERAPY
FR2760193B1 (en) 1997-02-28 1999-05-28 Transgene Sa LIPIDS AND COMPLEXES OF CATIONIC LIPIDS AND ACTIVE SUBSTANCES, IN PARTICULAR FOR THE TRANSFECTION OF CELLS
FR2763958A1 (en) 1997-05-29 1998-12-04 Transgene Sa COMBINATION PRODUCT COMBINING A NUCLEIC ACID WITH A SUBSTANCE DISORGANIZING THE EXTRACELLULAR MATRIX FOR GENE THERAPY
EP0989849A2 (en) 1997-06-13 2000-04-05 The Johns Hopkins University School Of Medicine Therapeutic nanospheres
AU707186B2 (en) 1997-07-01 1999-07-01 Transgene S.A. Compositions useful for transferring therapeutically active substances into a target cell, and their use in gene therapy
EP0974668B1 (en) 1998-07-07 2002-10-02 Transgene S.A. Use of adenoviral E4 reading frames to improve expression of a gene of interest
CA2364934C (en) 1999-02-22 2011-10-18 Transgene S.A. Method for obtaining a purified viral preparation
US6511832B1 (en) 1999-10-06 2003-01-28 Texas A&M University System In vitro synthesis of capped and polyadenylated mRNAs using baculovirus RNA polymerase
EP1278551A2 (en) 2000-04-21 2003-01-29 Vical Incorporated Compositions and methods for (in vivo) delivery of polynucleotide-based therapeutics
JP4588296B2 (en) * 2001-04-05 2010-11-24 ジョンズ・ホプキンス・ユニバーシティ Chimera vaccine
EP1795599A1 (en) * 2005-12-09 2007-06-13 Schuler, Gerold, Prof. Dr. Methods for generating antigen-specific effector T cells
US9085638B2 (en) * 2007-03-07 2015-07-21 The Johns Hopkins University DNA vaccine enhancement with MHC class II activators
EP2161279A1 (en) 2008-08-29 2010-03-10 Centre National de la Recherche Scientifique Synthetic peptides corresponding to overlapping neutralizing determinants in the CBD1 epitope induce broadly neutralizing antibodies
EP2470554B1 (en) 2009-08-26 2017-06-28 Selecta Biosciences, Inc. Compositions that induce t cell help
US20120294879A1 (en) 2009-10-13 2012-11-22 National University Of Singapore Consensus sequence for influenza a virus
WO2012140627A1 (en) * 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
DK2861240T3 (en) * 2012-06-15 2020-09-28 Immunomic Therapeutics Inc NUCLEIC ACIDS FOR THE TREATMENT OF ALLERGIES
CA3061206A1 (en) * 2017-04-22 2018-10-25 Immunomic Therapeutics, Inc. Improved lamp constructs

Also Published As

Publication number Publication date
IL270271B2 (en) 2023-03-01
JP2024037951A (en) 2024-03-19
CN110913892A (en) 2020-03-24
EP3618854A1 (en) 2020-03-11
IL270271A (en) 2019-12-31
JP7407902B2 (en) 2024-01-04
KR20240027895A (en) 2024-03-04
JP2023036863A (en) 2023-03-14
JP2020518613A (en) 2020-06-25
WO2018204534A1 (en) 2018-11-08
IL297073A (en) 2022-12-01
CA3061950A1 (en) 2018-11-08
BR112019023014A2 (en) 2020-05-19
IL297073B1 (en) 2023-08-01
KR20200016224A (en) 2020-02-14
AU2018263923A1 (en) 2019-10-31
US20200087365A1 (en) 2020-03-19
IL270271B (en) 2022-11-01
JP7222915B2 (en) 2023-02-15

Similar Documents

Publication Publication Date Title
US11773153B2 (en) LAMP constructs
JP7407902B2 (en) Improved LAMP constructs containing cancer antigens
US20240108703A1 (en) Improved LAMP Constructs Comprising Cancer Antigens
US20210261647A1 (en) Lamp constructs comprising allergens
JP2024056912A (en) Improved LAMP constructs
WO2023201201A1 (en) Bicistronic lamp constructs comprising immune response enhancing genes and methods of use thereof
Sas HER-2/neu-targeted immunoprevention of breast cancer